Language selection

Search

Patent 2754531 Summary

Third-party information liability

Some of the information on this Web page has been provided by external sources. The Government of Canada is not responsible for the accuracy, reliability or currency of the information supplied by external sources. Users wishing to rely upon this information should consult directly with the source of the information. Content provided by external sources is not subject to official languages, privacy and accessibility requirements.

Claims and Abstract availability

Any discrepancies in the text and image of the Claims and Abstract are due to differing posting times. Text of the Claims and Abstract are posted:

  • At the time the application is open to public inspection;
  • At the time of issue of the patent (grant).
(12) Patent Application: (11) CA 2754531
(54) English Title: ANTIBODY DRUG CONJUGATES (ADC) THAT BIND TO 24P4C12 PROTEINS
(54) French Title: CONJUGUES ANTICORPS-MEDICAMENT (ADC) SE LIANT AUX PROTEINES 24P4C12
Status: Dead
Bibliographic Data
(51) International Patent Classification (IPC):
  • C07K 19/00 (2006.01)
  • A61K 39/00 (2006.01)
  • A61P 35/00 (2006.01)
  • C07K 16/30 (2006.01)
  • C07K 14/705 (2006.01)
  • A61K 47/48 (2006.01)
(72) Inventors :
  • GUDAS, JEAN (United States of America)
  • JAKOBOVITS, AYA (United States of America)
  • AN, ZILI (United States of America)
  • MORRISON, ROBERT KENDALL (United States of America)
  • MORRISON, KAREN JANE MEYRICK (United States of America)
  • JIA, XIAO-CHI (United States of America)
  • BENJAMIN, DENNIS (United States of America)
  • MOSER, RUTH (United States of America)
  • SENTER, PETER (United States of America)
(73) Owners :
  • SEATTLE GENETICS, INC. (United States of America)
  • AGENSYS, INC. (United States of America)
(71) Applicants :
  • SEATTLE GENETICS, INC. (United States of America)
  • AGENSYS, INC. (United States of America)
(74) Agent: SMART & BIGGAR
(74) Associate agent:
(45) Issued:
(86) PCT Filing Date: 2010-03-05
(87) Open to Public Inspection: 2010-09-30
Availability of licence: N/A
(25) Language of filing: English

Patent Cooperation Treaty (PCT): Yes
(86) PCT Filing Number: PCT/US2010/026429
(87) International Publication Number: WO2010/111018
(85) National Entry: 2011-09-02

(30) Application Priority Data:
Application No. Country/Territory Date
61/158,143 United States of America 2009-03-06

Abstracts

English Abstract





Antibody drug conjugates (ADC's) that bind to 24P4C12 protein and variants
thereof are described herein.
24P4C12 exhibits tissue specific expression in normal adult tissue, and is
aberrantly expressed in the cancers listed in Table I.
Consequently, the ADC's of the invention provide a therapeutic composition for
the treatment of cancer.


French Abstract

L'invention concerne des conjugués anticorps-médicaments (ADC) qui se lient à la protéine 24P4C12 et leurs variants. La protéine 24P4C12 présente une expression tissulaire spécifique dans le tissu d'adulte normal, elle est en outre exprimé de manière aberrante dans les cancers répertoriés dans le Tableau I. En conséquence, les ADC selon l'invention offrent une composition thérapeutique pour le traitement de cancer.

Claims

Note: Claims are shown in the official language in which they were submitted.





Claims

1. An antibody drug conjugate comprising an antibody or antigen binding
fragment to monomethyl auristatin E (MMAE), wherein the antibody or fragment
comprises the heavy chain variable region consisting of the amino acid
sequence ranging
from 20th Q to the 143th S of SEQ ID NO: 20 and the light chain variable
consisting of the
amino acid sequence ranging from 23th D to the 130th R SEQ ID NO: 22.


2. An antibody drug conjugate comprising an antibody or fragment that
comprises the variable regions of the heavy chains and light chains of an
antibody
produced by a hybridoma deposited under American Type Culture Collection
(ATCC)
Accession No. PTA-8602 conjugated to monomethyl auristatin E (MMAE).


3. The antibody drug conjugate of claim 3, wherein the antibody comprises the
heavy chain and light chain of an antibody produced by a hybridoma deposited
under
A.T.C.C. Accession No.: PTA-8602.


4. The antibody drug conjugate of claim 1, wherein the antibody comprises
the heavy chain consisting of the amino acid sequence ranging from 20th Q to
the 469th K
of SEQ ID NO: 20 and the light chain consisting of the amino acid sequence
ranging from
23th D to the 236th C of SEQ ID NO: 22.


5. The antibody drug conjugate of claim 1, wherein the fragment is an Fab,
F(ab')2, Fv or Sfv fragment.


6. The antibody drug conjugate of claim 1, wherein the antibody is a fully
human antibody.


7. The antibody drug conjugate of claim 1, which the antibody is
recombinantly produced.


8. A pharmaceutical composition that comprises the antibody drug conjugate
of claim 1 in a human unit dose form.



117




9. The pharmaceutical composition of claim 8, wherein the composition is
for cancer treatment.


10. The pharmaceutical composition of claim 9, wherein the cancer is colon
cancer, pancreatic cancer, ovarian cancer, prostate cancer, or gastric cancer.


11. A method of inhibiting growth of cancer cells in a subject, comprising
administering to said subject an antibody drug conjugate of claim 1.


12. A method for treating tumor in a mammal comprising treating the mammal
with an effective amount of an antibody drug conjugate of claim 1.


13. A method for reducing tumor growth in a mammal comprising treating the
mammal with an effective amount of a combination of an antibody drug conjugate
and
radiation.


14. A method for reducing tumor growth in a mammal comprising treating the
mammal with an effective amount of a combination of an antibody drug conjugate
and a
chemotherapeutic agent.


15. A method for reducing tumor growth in a mammal comprising treating the
mammal with an effective amount of a combination of an antibody drug conjugate
and a
drug or biologically active therapy.



118

Description

Note: Descriptions are shown in the official language in which they were submitted.



CA 02754531 2011-09-02
WO 2010/111018 PCT/US2010/026429
ANTIBODY DRUG CONJUGATES (ADC)
THAT BIND TO 24P4C12 PROTEINS
STATEMENT OF RIGHTS TO INVENTIONS MADE UNDER FEDERALLY
SPONSORED RESEARCH

[0001] Not applicable.
FIELD OF THE INVENTION

[0002] The invention described herein relates to antibodies, binding
fragments, and
antibody drug conjugates (ADCs) thereof, that bind proteins, termed 24P4C12.
The
invention further relates to prognostic, prophylactic and therapeutic methods
and
compositions useful in the treatment of cancers that express 24P4C12.
BACKGROUND OF THE INVENTION

[0003] Cancer is the second leading cause of human death next to coronary
disease.
Worldwide, millions of people die from cancer every year. In the United States
alone, as
reported by the American Cancer Society, cancer causes the death of well over
a half-
million people annually, with over 1.2 million new cases diagnosed per year.
While
deaths from heart disease have been declining significantly, those resulting
from cancer
generally are on the rise. In the early part of the next century, cancer is
predicted to
become the leading cause of death.
[0004] Worldwide, several cancers stand out as the leading killers. In
particular,
carcinomas of the lung, prostate, breast, colon, pancreas, ovary, and bladder
represent the
primary causes of cancer death. These and virtually all other carcinomas share
a common
lethal feature. With very few exceptions, metastatic disease from a carcinoma
is fatal.
Moreover, even for those cancer patients who initially survive their primary
cancers,
common experience has shown that their lives are dramatically altered. Many
cancer
patients experience strong anxieties driven by the awareness of the potential
for
recurrence or treatment failure. Many cancer patients experience physical
debilitations
following treatment. Furthermore, many cancer patients experience a
recurrence.


CA 02754531 2011-09-02
WO 2010/111018 PCT/US2010/026429
[0005] Worldwide, prostate cancer is the fourth most prevalent cancer in men.
In
North America and Northern Europe, it is by far the most common cancer in
males and is
the second leading cause of cancer death in men. In the United States alone,
well over
30,000 men die annually of this disease - second only to lung cancer. Despite
the
magnitude of these figures, there is still no effective treatment for
metastatic prostate
cancer. Surgical prostatectomy, radiation therapy, hormone ablation therapy,
surgical
castration and chemotherapy continue to be the main treatment modalities.
Unfortunately, these treatments are ineffective for many and are often
associated with
undesirable consequences.
[0006] On the diagnostic front, the lack of a prostate tumor marker that can
accurately
detect early-stage, localized tumors remains a significant limitation in the
diagnosis and
management of this disease. Although the serum prostate specific antigen (PSA)
assay
has been a very useful tool, its specificity and general utility is widely
regarded as lacking
in several important respects.
[0007] Progress in identifying additional specific markers for prostate cancer
has been
improved by the generation of prostate cancer xenografts that can recapitulate
different
stages of the disease in mice. The LAPC (Los Angeles Prostate Cancer)
xenografts are
prostate cancer xenografts that have survived passage in severe combined
immune
deficient (SCID) mice and have exhibited the capacity to mimic the transition
from
androgen dependence to androgen independence (Klein et al., 1997, Nat. Med.
3:402).
More recently identified prostate cancer markers include PCTA-1 (Su et al.,
1996, Proc.
Natl. Acad. Sci. USA 93: 7252), prostate-specific membrane antigen (PSMA)
(Pinto
et al., Clin Cancer Res 1996 Sep 2 (9): 1445-51), STEAP (Hubert, et al., Proc
Natl Acad
Sci U S A. 1999 Dec 7; 96(25): 14523-8) and prostate stem cell antigen (PSCA)
(Reiter
et al., 1998, Proc. Natl. Acad. Sci. USA 95: 1735).
[0008] While previously identified markers such as PSA have facilitated
efforts to
diagnose and treat prostate cancer, there is need for the identification of
additional
markers and therapeutic targets for prostate and related cancers in order to
further
improve diagnosis and therapy. An estimated 130,200 cases of colorectal cancer
occurred
in 2000 in the United States, including 93,800 cases of colon cancer and
36,400 of rectal
cancer.

2


CA 02754531 2011-09-02
WO 2010/111018 PCT/US2010/026429
[0009] Colorectal cancers are the third most common cancers in men and women.
Incidence rates declined significantly during 1992-1996 (-2.1% per year).
Research
suggests that these declines have been due to increased screening and polyp
removal,
preventing progression of polyps to invasive cancers. There were an estimated
56,300
deaths (47,700 from colon cancer, 8,600 from rectal cancer) in 2000,
accounting for about
11% of all U.S. cancer deaths.
[0010] At present, surgery is the most common form of therapy for colorectal
cancer,
and for cancers that have not spread, it is frequently curative. Chemotherapy,
or
chemotherapy plus radiation, is given before or after surgery to most patients
whose
cancer has deeply perforated the bowel wall or has spread to the lymph nodes.
A
permanent colostomy (creation of an abdominal opening for elimination of body
wastes)
is occasionally needed for colon cancer and is infrequently required for
rectal cancer.
There continues to be a need for effective diagnostic and treatment modalities
for
colorectal cancer.
[0011] Of all new cases of cancer in the United States, bladder cancer
represents
approximately 5 percent in men (fifth most common neoplasm) and 3 percent in
women
(eighth most common neoplasm). The incidence is increasing slowly, concurrent
with an
increasing older population. In 1998, there were an estimated 54,500 cases,
including
39,500 in men and 15,000 in women. The age-adjusted incidence in the United
States is
32 per 100,000 for men and eight per 100,000 in women. The historic
male/female ratio
of 3:1 may be decreasing related to smoking patterns in women. There were an
estimated
11,000 deaths from bladder cancer in 1998 (7,800 in men and 3,900 in women).
Bladder
cancer incidence and mortality strongly increase with age and will be an
increasing
problem as the population becomes more elderly.
[0012] Most bladder cancers recur in the bladder. Bladder cancer is managed
with a
combination of transurethral resection of the bladder (TUR) and intravesical
chemotherapy or immunotherapy. The multifocal and recurrent nature of bladder
cancer
points out the limitations of TUR. Most muscle-invasive cancers are not cured
by TUR
alone. Radical cystectomy and urinary diversion is the most effective means to
eliminate
the cancer but carry an undeniable impact on urinary and sexual function.
There
continues to be a significant need for treatment modalities that are
beneficial for bladder
cancer patients.

3


CA 02754531 2011-09-02
WO 2010/111018 PCT/US2010/026429
[0013] There were an estimated 164,100 new cases of lung and bronchial cancer
in
2000, accounting for 14% of all U.S. cancer diagnoses. The incidence rate of
lung and
bronchial cancer is declining significantly in men, from a high of 86.5 per
100,000 in
1984 to 70.0 in 1996. In the 1990s, the rate of increase among women began to
slow. In
1996, the incidence rate in women was 42.3 per 100,000.
[0014] Lung and bronchial cancer caused an estimated 156,900 deaths in 2000,
accounting for 28% of all cancer deaths. During 1992-1996, mortality from lung
cancer
declined significantly among men (-1.7% per year) while rates for women were
still
significantly increasing (0.9% per year). Since 1987, more women have died
each year of
lung cancer than breast cancer, which, for over 40 years, was the major cause
of cancer
death in women. Decreasing lung cancer incidence and mortality rates most
likely
resulted from decreased smoking rates over the previous 30 years; however,
decreasing
smoking patterns among women lag behind those of men. Of concern, although the
declines in adult tobacco use have slowed, tobacco use in youth is increasing
again.
[0015] Treatment options for lung and bronchial cancer are determined by the
type
and stage of the cancer and include surgery, radiation therapy, and
chemotherapy. For
many localized cancers, surgery is usually the treatment of choice. Because
the disease
has usually spread by the time it is discovered, radiation therapy and
chemotherapy are
often needed in combination with surgery. Chemotherapy alone or combined with
radiation is the treatment of choice for small cell lung cancer; on this
regimen, a large
percentage of patients experience remission, which in some cases is long
lasting. There is
however, an ongoing need for effective treatment and diagnostic approaches for
lung and
bronchial cancers.
[0016] An estimated 182,800 new invasive cases of breast cancer were expected
to
occur among women in the United States during 2000. Additionally, about 1,400
new
cases of breast cancer were expected to be diagnosed in men in 2000. After
increasing
about 4% per year in the 1980s, breast cancer incidence rates in women have
leveled off
in the 1990s to about 110.6 cases per 100,000.
[0017] In the U.S. alone, there were an estimated 41,200 deaths (40,800 women,
400
men) in 2000 due to breast cancer. Breast cancer ranks second among cancer
deaths in
women. According to the most recent data, mortality rates declined
significantly during
4


CA 02754531 2011-09-02
WO 2010/111018 PCT/US2010/026429
1992-1996 with the largest decreases in younger women, both white and black.
These
decreases were probably the result of earlier detection and improved
treatment.
[0018] Taking into account the medical circumstances and the patient's
preferences,
treatment of breast cancer may involve lumpectomy (local removal of the tumor)
and
removal of the lymph nodes under the arm; mastectomy (surgical removal of the
breast)
and removal of the lymph nodes under the arm; radiation therapy; chemotherapy;
or
hormone therapy. Often, two or more methods are used in combination. Numerous
studies have shown that, for early stage disease, long-term survival rates
after
lumpectomy plus radiotherapy are similar to survival rates after modified
radical
mastectomy. Significant advances in reconstruction techniques provide several
options
for breast reconstruction after mastectomy. Recently, such reconstruction has
been done
at the same time as the mastectomy.
[0019] Local excision of ductal carcinoma in situ (DCIS) with adequate amounts
of
surrounding normal breast tissue may prevent the local recurrence of the DCIS.
Radiation to the breast and/or tamoxifen may reduce the chance of DCIS
occurring in the
remaining breast tissue. This is important because DCIS, if left untreated,
may develop
into invasive breast cancer. Nevertheless, there are serious side effects or
sequelae to
these treatments. There is, therefore, a need for efficacious breast cancer
treatments.
[0020] There were an estimated 23,100 new cases of ovarian cancer in the
United
States in 2000. It accounts for 4% of all cancers among women and ranks second
among
gynecologic cancers. During 1992-1996, ovarian cancer incidence rates were
significantly declining. Consequent to ovarian cancer, there were an estimated
14,000
deaths in 2000. Ovarian cancer causes more deaths than any other cancer of the
female
reproductive system.
[0021] Surgery, radiation therapy, and chemotherapy are treatment options for
ovarian cancer. Surgery usually includes the removal of one or both ovaries,
the fallopian
tubes (salpingo-oophorectomy), and the uterus (hysterectomy). In some very
early
tumors, only the involved ovary will be removed, especially in young women who
wish
to have children. In advanced disease, an attempt is made to remove all intra-
abdominal
disease to enhance the effect of chemotherapy. There continues to be an
important need
for effective treatment options for ovarian cancer.



CA 02754531 2011-09-02
WO 2010/111018 PCT/US2010/026429
[0022] There were an estimated 28,300 new cases of pancreatic cancer in the
United
States in 2000. Over the past 20 years, rates of pancreatic cancer have
declined in men.
Rates among women have remained approximately constant but may be beginning to
decline. Pancreatic cancer caused an estimated 28,200 deaths in 2000 in the
United
States. Over the past 20 years, there has been a slight but significant
decrease in mortality
rates among men (about -0.9% per year) while rates have increased slightly
among
women.
[0023] Surgery, radiation therapy, and chemotherapy are treatment options for
pancreatic cancer. These treatment options can extend survival and/or relieve
symptoms
in many patients but are not likely to produce a cure for most. There is a
significant need
for additional therapeutic and diagnostic options for cancers. These include
the use of
antibodies, vaccines, and small molecules as treatment modalities.
Additionally, there is
also a need to use these modilities as research tools to diagnose, detect,
monitor, and
further the state of the art in all areas of cancer treatment and studies.
[0024] The therapeutic utility of monoclonal antibodies (mAbs) (G. Kohler and
C.
Milstein, Nature 256:495-497 (1975)) is being realized. Monoclonal antibodies
have now
been approved as therapies in transplantation, cancer, infectious disease,
cardiovascular
disease and inflammation. Different isotypes have different effector
functions. Such
differences in function are reflected in distinct 3-dimensional structures for
the various
immunoglobulin isotypes (P.M. Alzari et al., Annual Rev. Immunol., 6:555-580
(1988)).
[0025] Because mice are convenient for immunization and recognize most human
antigens as foreign, mAbs against human targets with therapeutic potential
have typically
been of murine origin. However, murine mAbs have inherent disadvantages as
human
therapeutics. They require more frequent dosing as mAbs have a shorter
circulating half-
life in humans than human antibodies. More critically, the repeated
administration of
murine antibodies to the human immune system causes the human immune system to
respond by recognizing the mouse protein as a foreign and generating a human
anti-
mouse antibody (HAMA) response. Such a HAMA response may result in allergic
reaction and the rapid clearing of the murine antibody from the system thereby
rendering
the treatment by murine antibody useless. To avoid such affects, attempts to
create
human immune systems within mice have been attempted.

6


CA 02754531 2011-09-02
WO 2010/111018 PCT/US2010/026429
[0026] Initial attempts hoped to create transgenic mice capable of responding
to
antigens with antibodies having human sequences (See Bruggemann et al., Proc.
Nat'l.
Acad. Sci. USA 86:6709-6713 (1989)), but were limited by the amount of DNA
that
could be stably maintained by available cloning vehicles. The use of yeast
artificial
chromosome (YAC) cloning vectors led the way to introducing large germline
fragments
of human Ig locus into transgenic mammals. Essentially a majority of the human
V, D,
and J region genes arranged with the same spacing found in the human genome
and the
human constant regions were introduced into mice using YACs. One such
transgenic
mouse strain is known as XenoMouse mice and is commercially available from
Amgen
Fremont, Inc. (Fremont CA).

SUMMARY OF THE INVENTION

[0027] The invention provides antibodies, binding fragments, and antibody drug
conjugates (ADCs) thereof that bind to 24P4C12 proteins and polypeptide
fragments of
24P4C12 proteins. In some embodiments, the invention comprises fully human
antibodies conjugated with a therapeutic agent. In certain embodiments, there
is a
proviso that the entire nucleic acid sequence of Figure 3 is not encoded
and/or the entire
amino acid sequence of Figure 2 is not prepared. In certain embodiments, the
entire
nucleic acid sequence of Figure 3 is encoded and/or the entire amino acid
sequence of
Figure 2 is prepared, either of which are in respective human unit dose forms.
[0028] The invention further provides various immunogenic or therapeutic
compositions, such as antibody drug conjugates, and strategies for treating
cancers that
express 24P4C12 such as cancers of tissues listed in Table I.

BRIEF DESCRIPTION OF THE FIGURES

[0029] Figure 1. Nucleic Acid and Amino Acid Sequences of 24P4C12. Figure
1A. The cDNA and amino acid sequence of 24P4C12 variant 1 (also called
"24P4C12
v.1" or "24P4C12 variant 1") is shown in Figure 1A. The start methionine is
underlined.
The open reading frame extends from nucleic acid 6-2138 including the stop
codon.
[0030] Figure 1B. The cDNA and amino acid sequence of 24P4C12 variant 2 (also
called "24P4C12 v.2") is shown in Figure 1B. The codon for the start
methionine is

7


CA 02754531 2011-09-02
WO 2010/111018 PCT/US2010/026429
underlined. The open reading frame extends from nucleic acid 6-2138 including
the stop
codon.
[0031] Figure 1C. The cDNA and amino acid sequence of 24P4C12 variant 3 (also
called "24P4C12 v.3") is shown in Figure 1C. The codon for the start
methionine is
underlined. The open reading frame extends from nucleic acid 6-2138 including
the stop
codon.
[0032] Figure 1D. The cDNA and amino acid sequence of 24P4C12 variant 4 (also
called "24P4C12 v.4") is shown in Figure 1D. The codon for the start
methionine is
underlined. The open reading frame extends from nucleic acid 6-2138 including
the stop
codon.
[0033] Figure 1E. The cDNA and amino acid sequence of 24P4C12 variant 5 (also
called "24P4C12 v.5") is shown in Figure 1E. The codon for the start
methionine is
underlined. The open reading frame extends from nucleic acid 6-2138 including
the stop
codon.
[0034] Figure 1F. The cDNA and amino acid sequence of 24P4C12 variant 6 (also
called "24P4C12 v.6") is shown in Figure IF. The codon for the start
methionine is
underlined. The open reading frame extends from nucleic acid 6-2138 including
the stop
codon.
[0035] Figure 1G. The cDNA and amino acid sequence of 24P4C12 variant 7 (also
called "24P4C12 v.7") is shown in Figure 1G. The codon for the start
methionine is
underlined. The open reading frame extends from nucleic acid 6-1802 including
the stop
codon.
[0036] Figure M. The cDNA and amino acid sequence of 24P4C12 variant 8 (also
called "24P4C12 v.8") is shown in Figure 1H. The codon for the start
methionine is
underlined. The open reading frame extends from nucleic acid 6-2174 including
the stop
codon.
[0037] Figure 11. The cDNA and amino acid sequence of 24P4C12 variant 9 (also
called "24P4C12 v.9") is shown in Figure 11. The codon for the start
methionine is
underlined. The open reading frame extends from nucleic acid 6-2144 including
the stop
codon.
[0038] Figure 2. Nucleic Acid and Amino Acid sequences of 24P4C12 antibodies.
8


CA 02754531 2011-09-02
WO 2010/111018 PCT/US2010/026429
[0039] Figure 2A. The cDNA and amino acid sequence of Ha5-1(5)2.1 heavy chain.
Double-underlined is the leader sequence, underlined is the heavy chain
variable region,
and underlined with a dashed line is the human IgG2 constant region.
[0040] Figure 2B. The cDNA and amino acid sequence of Ha5-1(5)2.1 light chain.
Double-underlined is the leader sequence, underlined is the light chain
variable region,
and underlined with a dashed line is the human kappa constant region.
[0041] Figure 3. Amino Acid sequences of 24P4C12 antibodies.
[0042] Figure 3A. The amino acid sequence of Ha5-1(5)2.1 heavy chain. Double-
underlined is the leader sequence, underlined is the heavy chain variable
region, and
underlined with a dashed line is the human IgG2 constant region.
[0043] Figure 3B. The amino acid sequence of Ha5-1(5)2.1 light chain. Double-
underlined is the leader sequence, underlined is the light chain variable
region, and
underlined with a dashed line is the human kappa constant region.
[0044] Figure 4. Alignment of Ha5-1(5)2.1 antibodies to human Ig germline.
[0045] Figure 4A. Alignment of Ha5-1(5)2.1 heavy chain to human Ig germline.
[0046] Figure 4B. Alignment of Ha5-1(5)2.1 light chain to human Ig germline.
[0047] Figure 5. Ha5-1(5)2.1 MAb binds to cell surface of 24P4C12. PC3-control
and PC3-24P4C12 cells were stained with Ha5-1(5)2.1 MAb purified from either
hybridoma or from CHO cells transfected with Ha5-1(5)2.1 heavy and light chain
vector
constructs. Binding was detected by flow cytometry. Results show Ha5-1(5)2.1
produced by CHO cells bind 24P4C12 similarly to the Ha5-1(5)2.1 hybridoma
product.
[0048] Figure 6. Cell Cytotoxicity by Ha5-1(5)2.1-vcMMAE. Cytotoxicity by Ha5-
1(5)2.1 -vcMMAE was evaluated in PC3 cells engineered to express 24P4C12. PC3-
Neo
or PC3-24P4C12 cells (1000 cells/well) were seeded into a 96 well plate on day
1. The
following day an equal volume of medium containing the indicated concentration
of
Ha5-1(5)2.1-vcMMAE or a Control MAb conjugated with vc-MMAE was added to each
well. The cells were allowed to incubate for 4 days at 37 degrees C. At the
end of the
incubation period, Alamar Blue was added to each well and incubation continued
for an
additional 4 hours. The resulting fluorescence was detected using a Biotek
plate reader
with an excitation wavelength of 620 nm and an emission wavelength of 540 nm.
The
results in show that Ha5-1(5)2.1-vcMMAE mediated cytotoxicity in PC3-24P4C12
cells
while a control human IgG conjugated with vcMMAE had no effect. These results

9


CA 02754531 2011-09-02
WO 2010/111018 PCT/US2010/026429
indicate that Ha5-1(5)2.1-vcMMAE can selectively deliver a cytotoxic drug to
24P4C12
expressing cells leading to their killing.
[0049] Figure 7. Ha5-1(5)2.1vcMMAE inhibits the growth of subcutaneous
established human androgen-independent prostate cancer xenograft in SCID mice.
In this
experiment, androgen-independent human prostate cancer PC-3-Hu24P4C12 tumor
cells
(3.0 x 106 cells/mouse) were injected subcutaneously into male SCID mice. Mice
were
randomized into Ha5-1(5)2.1-vcMMAE and PBS control groups (n=5 in each group)
when tumors reached 100 mm3. Mice were treated with a single dose of Ha5-
1(5)2.1-
vcMMAE (10 mg/kg) or PBS administered intravenously (i.v.) on Day 0. Tumor
growth
was monitored using caliper measurements every 3 to 4 days as indicated. Tumor
volume
was calculated as Width2 x Length/2, where width is the smallest dimension and
length is
the largest. The results show that treatment with Ha5-1(5)2.1-vcMMAE
significantly
inhibited the growth of PC-3-Hu24P4C12 prostate tumors in SCID mice (p< 0.01)
and
resulted in complete tumor regression in most animals.
[0050] Figure 8. Ha5-1(5)2.1vcMMAE inhibits the growth of orthotopically
established human androgen-independent prostate cancer xenograft in SCID mice.
LAPC-9AI androgen-independent human prostate cancer cells (2.0 x 106
cells/mouse)
were implanted into the prostates of male SCID mice. Fifteen (15) days after
implantation
when tumors were well established and palpable, the mice were randomized into
two
groups (n=8 in each group). Mice were treated with either Ha5-1(5)2.1-vcMMAE
or
isotype control MAb conjugated with vcMMAE administered i.v. at 3 mg/kg every
4 days
for a total of 4 doses. At the end of study tumors in the mouse prostate were
excised and
weighed using an electronic balance. The results show that treatment with Ha5-
1(5)2.1-
vcMMAE significantly inhibited the growth of LAPC9-AI human prostate tumors
implanted orthotopically in SCID mice (p< 0.01).
[0051] Figure 9. Ha5-1(5)2.1vcMMAE inhibits the growth of subcutaneous
established human androgen-independent human colon cancer xenograft in SCID
mice.
HT-29 human colon cancer cells (1.0 x 106 cells/mouse) were injected
subcutaneously
into SCID mice. Mice were randomized into two groups (n=6 in each group) when
tumors reached 100 mm3. Ha5-1(5)2.1-vcMMAE (3 mg/kg) or PBS was administered
intravenously every 4 days for a total of 4 doses beginning on Day 0. Tumor
growth was
monitored using caliper measurements every 3 to 4 days as indicated. Tumor
volume was



CA 02754531 2011-09-02
WO 2010/111018 PCT/US2010/026429
calculated as Width2 x Length/2, where width is the smallest dimension and
length is the
largest. The results show that treatment with Ha5-1(5)2.1-vcMMAE significantly
inhibited the growth of HT-29 human colon tumor xenografts implanted
subcutaneously
in SCID mice (p< 0.01).
[0052] Figure 10. Ha5-1(5)2.1vcMMAE inhibits the growth of subcutaneous
established patient-derived colon cancer xenograft in SCID mice. AG-C4,
patient-
derived colon cancer xenograft tumor pieces, were implanted subcutaneously
into SCID
mice. Mice were randomized into two groups (n=6 in each group) when tumors
reached
100 mm3. Ha5-1(5)2.1-vcMMAE (3 mg/kg ) or PBS was administered intravenously
every 3-4 days for a total of 4 doses starting on Day 0. Tumor growth was
monitored
using caliper measurements every 3 to 4 days as indicated. Tumor volume was
calculated
as Width2 x Length/2, where width is the smallest dimension and length is the
largest.
The results show that treatment with Ha5-1(5)2.1-vcMMAE significantly
inhibited the
growth of AG-C4 human colon tumor xenografts implanted subcutaneously in SCID
mice
(p< 0.05).
[0053] Figure 11. Ha5-1(5)2.1vcMMAE inhibits the growth of subcutaneous
established human ovarian cancer xenograft in nude mice. OVCAR-5 human ovarian
cancer tumor cells (2.0 x 106 cells/mouse) were injected subcutaneously into
the nude
mice. Mice were randomized into two groups (n=6 in each group) when tumors
reached
100 mm3. Ha5-1(5)2.1-vcMMAE (5 mg/kg) or PBS was administered intravenously
once every 3-4 days for a total of 4 doses starting on Day 0. Tumor growth was
monitored
using caliper measurements every 3 to 4 days as indicated. Tumor volume was
calculated
as Width2 x Length/2, where width is the smallest dimension and length is the
largest.
The results show that treatment with Ha5-1(5)2.1-vcMMAE significantly
inhibited the
growth of OVCAR-5 ovarian cancer xenografts implanted subcutaneously in nude
mice
(p< 0.01).
[0054] Figure 12. Ha5-1(5)2.1vcMMAE inhibits the growth of subcutaneous
established patient-derived pancreatic cancer xenograft in SCID mice. AG-Panc3
patient-
derived pancreatic tumor pieces were implanted subcutaneously into SCID mice.
Mice
were randomized into two groups (n=6 in each group) when tumors reached 85
mm3.
Ha5-1(5)2.1-vcMMAE (5 mg/kg) or PBS was administered intravenously once every
3-4
days for a total of 4 doses beginning on Day 0. Tumor growth was monitored
using

11


CA 02754531 2011-09-02
WO 2010/111018 PCT/US2010/026429
caliper measurements every 3 to 4 days as indicated. Tumor volume was
calculated as
Width2 x Length/2, where width is the smallest dimension and length is the
largest. The
results show that treatment with Ha5-1(5)2.1-vcMMAE significantly inhibited
the growth
of AG-Panc3 tumor xenografts implanted subcutaneously in SCID mice (p< 0.01).
[0055] Figure 13. Efficacy of Ha5-1(5)2.1vcMMAE compared to other 24P4C12
Antibody Drug Conjugates (ADCs) in Prostate Cancer LAPC9-AD Xenografts. LAPC-
9AD androgen-dependent human prostate cancer cells (1.5 x 106 cells/mouse)
were
injected subcutaneously into male SCID mice. Mice were randomized into Ha5-
1(5)2.1-
vcMMAE, Ha5-1(5)2.1-mcMMAF and other Antibody Drug Conjugate (ADC) groups
including a PBS control group (n=6 in each group), as shown in graph (Figure
13). When
tumors reached 100 mm3, Ha5-1(5)2.1-vcMMAE, Ha5-1(5)2.1-mcMMAF and all other
ADCs were administered intravenously at 10 mg/kg once on day 0. Tumor growth
was
monitored using caliper measurements every 3 to 4 days as indicated. Tumor
volume was
calculated as Width2 x Length/2, where width is the smallest dimension and
length is the
largest. The results show that treatment with Ha5-1(5)2.1-vcMMAE significantly
inhibited the growth of LAPC9-AD prostate cancer xenografts as compared to Ha5-

1(5)2.1-mcMMAF (p=0.0048). (Figure 13). Other antibodies conjugated to -vcMMAE
and -mcMMAF did not have any tumor inhibitory activity which shows that Ha5-
1(5)2.1
posesses a significant prominent effect of inhibiting tumor growth and can be
used for
therapeutic purposes to treat and manage cancers set forth in Table I.
[0056] Figure 14. Detection of 24P4C12 protein in gastric cancer patient
specimens
by IHC. Expression of 24P4C12 protein by immunohistochemistry was tested in
two (2)
different tumor specimens from gastric cancer patients. Briefly, formalin
fixed, paraffin
wax-embedded tissues were cut into 4 micron sections and mounted on glass
slides. The
sections were de-waxed, rehydrated and treated with trypsin solution (0.05%
trypsin
(ICN, Aurora, Ohio) in 0.05% calcium chloride, with pH adjusted to 7.8) at 37
C for 10
minutes. Sections were then treated with 3% hydrogen peroxide solution to
inactivate
endogenous peroxidase activity. Serum-free protein block (Dako, Carpenteria,
CA) was
used to inhibit non-specific binding prior to incubation with monoclonal mouse
anti-
24P4C12 antibody or an isotype control. Subsequently, the sections were
treated with the
Super SensitiveTM Polymer-horseradish peroxidase (HRP) Detection System which
consists of an incubation in Super EnhancerTM reagent followed by an
incubation with

12


CA 02754531 2011-09-02
WO 2010/111018 PCT/US2010/026429
polymer-HRP secondary antibody conjugate (BioGenex, San Ramon, CA). The
sections
were then developed using the DAB kit (BioGenex, San Ramon, CA), nuclei were
stained
using hematoxylin, and analyzed by bright field microscopy. Specific staining
was
detected in patient specimens using the 24P4C12 immunoreactive antibody, as
indicated
by the brown staining. (See, Figures 14(A) and 14(C). In contrast, the control
antibody
did not stain either patient specimen. (See, Figures 14(B) and 14(D). The
results show
expression of 24P4C12 in the tumor cells of patient gastric cancer tissues.
These results
indicate that 24P4C12 is expressed in human cancers and that antibodies
directed to this
antigen (e.g. Ha5-1(5)2.1) are useful for diagnostic and therapeutic purposes.
(Figure
14(A) - 14(D)).

DETAILED DESCRIPTION OF THE INVENTION
Outline of Sections
I.) Definitions
II.) 24P4C12 Antibodies
III.) Antibody Drug Conjugates Generally
III(A). Maytansinoids
III(B). Auristatins and dolostatins
III(C). Calicheamicin
III(D). Other Cytotoxic Agents
IV.) Antibody Drug Conjugates which Bind 24P4C12
V.) Linker Units
VI.) The Stretcher Unit
VII.) The Amino Acid Unit
VIII.) The Spacer Unit
IX.) The Drug Unit
X.) Drug Loading
XI.) Methods of Determining Cytotoxic effect of ADCs
XII.) Treatment of Cancer(s) Expressing 24P4C12
XIII.) 24P4C12 as a Target for Antibody-based Therapy
XIV.) 24P4C12 ADC Cocktails
XV.) Combination Therapy
XVI.) KITS/Articles of Manufacture
I.) Definitions:
[0057] Unless otherwise defined, all terms of art, notations and other
scientific terms
or terminology used herein are intended to have the meanings commonly
understood by
those of skill in the art to which this invention pertains. In some cases,
terms with
commonly understood meanings are defined herein for clarity and/or for ready
reference,

13


CA 02754531 2011-09-02
WO 2010/111018 PCT/US2010/026429
and the inclusion of such definitions herein should not necessarily be
construed to
represent a substantial difference over what is generally understood in the
art. Many of
the techniques and procedures described or referenced herein are well
understood and
commonly employed using conventional methodology by those skilled in the art,
such as,
for example, the widely utilized molecular cloning methodologies described in
Sambrook
et al., Molecular Cloning: A Laboratory Manual 2nd. edition (1989) Cold Spring
Harbor
Laboratory Press, Cold Spring Harbor, N.Y. As appropriate, procedures
involving the
use of commercially available kits and reagents are generally carried out in
accordance
with manufacturer defined protocols and/or parameters unless otherwise noted.
[0058] When a trade name is used herein, reference to the trade name also
refers to
the product formulation, the generic drug, and the active pharmaceutical
ingredient(s) of
the trade name product, unless otherwise indicated by context.
[0059] The terms "advanced cancer", "locally advanced cancer", "advanced
disease"
and "locally advanced disease" mean cancers that have extended through the
relevant
tissue capsule, and are meant to include stage C disease under the American
Urological
Association (AUA) system, stage Cl - C2 disease under the Whitmore-Jewett
system, and
stage T3 - T4 and N+ disease under the TNM (tumor, node, metastasis) system.
In
general, surgery is not recommended for patients with locally advanced
disease, and these
patients have substantially less favorable outcomes compared to patients
having clinically
localized (organ-confined) cancer.
[0060] The abbreviation "AFP" refers to dimethylvaline-valine-dolaisoleuine-
dolaproine-phenylalanine-p-phenylenediamine (see Formula XVI infra).

[0061] The abbreviation "MMAE" refers to monomethyl auristatin E (see Formula
XI
infra).
[0062] The abbreviation "AEB" refers to an ester produced by reacting
auristatin E
with paraacetyl benzoic acid (see Formula XX infra).
[0063] The abbreviation "AEVB" refers to an ester produced by reacting
auristatin E
with benzoylvaleric acid (see Formula XXI infra).
[0064] The abbreviation "MMAF" refers to dovaline-valine-dolaisoleuine-
dolaproine-phenylalanine (see Formula XVIV infra).
[0065] Unless otherwise noted, the term "alkyl" refers to a saturated straight
or
branched hydrocarbon having from about 1 to about 20 carbon atoms (and all

14


CA 02754531 2011-09-02
WO 2010/111018 PCT/US2010/026429
combinations and subcombinations of ranges and specific numbers of carbon
atoms
therein), with from about 1 to about 8 carbon atoms being preferred. Examples
of alkyl
groups are methyl, ethyl, n-propyl, iso-propyl, n-butyl, iso-butyl, sec-butyl,
tert-butyl, n-
pentyl, 2-pentyl, 3-pentyl, 2-methyl-2-butyl, n-hexyl, n-heptyl, n-octyl, n-
nonyl, n-decyl,
3-methyl-2-butyl, 3-methyl-l-butyl, 2-methyl-l-butyl, 1-hexyl, 2-hexyl, 3-
hexyl, 2-
methyl-2-pentyl, 3-methyl-2-pentyl, 4-methyl-2-pentyl, 3-methyl-3-pentyl, 2-
methyl-3-
pentyl, 2,3-dimethyl-2-butyl, and 3,3-dimethyl-2-butyl.
[0066] Alkyl groups, whether alone or as part of another group, can be
optionally
substituted with one or more groups, preferably 1 to 3 groups (and any
additional
substituents selected from halogen), including, but not limited to, -halogen, -
O-(C1-C8
alkyl), -O-(C2-C8 alkenyl), -O-(C2-C8 alkynyl), -aryl, -C(O)R', -OC(O)R', -
C(O)OR', -
C(O)NH2, -C(O)NHR',
-C(O)N(R')2, -NHC(O)R', -SR', -SO3R', -S(O)2R', -S(O)R', -OH, =0, -N3 , -NH2, -

NH(R'),
-N(R')2 and -CN, where each R' is independently selected from -H, -CI-C8
alkyl, -C2-C8
alkenyl, -C2-C8 alkynyl, or -aryl, and wherein said -O-(C1-C8 alkyl), -O-(C2-
C8 alkenyl),
-O-(C2-C8 alkynyl), -aryl, -CI-C8 alkyl, -C2-C8 alkenyl, and -C2-C8 alkynyl
groups can be
optionally further substituted with one or more groups including, but not
limited to, -Ci-
C8 alkyl, -C2-C8 alkenyl, -C2-C8 alkynyl, -halogen, -O-(C1-C8 alkyl), -O-(C2-
C8 alkenyl), -
O-(C2-C8 alkynyl), -aryl, -C(O)R", -OC(O)R", -C(O)OR", -C(O)NH2, -C(O)NHR", -
C(O)N(R")2, -NHC(O)R", -SR", -SO3R", -S(O)2R", -S(O)R", -OH, -N3, -NH2, -
NH(R"), -N(R")2 and -CN, where each R" is independently selected from -H, -C1-
C8
alkyl, -C2-C8 alkenyl, -C2-C8 alkynyl, or
-aryl.
[0067] Unless otherwise noted, the terms "alkenyl" and "alkynyl" refer to
straight and
branched carbon chains having from about 2 to about 20 carbon atoms (and all
combinations and subcombinations of ranges and specific numbers of carbon
atoms
therein), with from about 2 to about 8 carbon atoms being preferred. An
alkenyl chain
has at least one double bond in the chain and an alkynyl chain has at least
one triple bond
in the chain. Examples of alkenyl groups include, but are not limited to,
ethylene or
vinyl, allyl, -1-butenyl, -2-butenyl, -isobutylenyl,
-1-pentenyl, -2-pentenyl, -3-methyl-l-butenyl, -2-methyl-2-butenyl, and -


CA 02754531 2011-09-02
WO 2010/111018 PCT/US2010/026429
2,3-dimethyl-2- butenyl. Examples of alkynyl groups include, but are not
limited to,
acetylenic, propargyl, acetylenyl, propynyl, -1-butynyl, -2-butynyl, -1-
pentynyl, -
2-pentynyl, and
-3-methyl-1 butynyl.
[0068] Alkenyl and alkynyl groups, whether alone or as part of another group,
can be
optionally substituted with one or more groups, preferably 1 to 3 groups (and
any
additional substituents selected from halogen), including but not limited to, -
halogen, -0-
(C1-C8 alkyl), -O-(C2-C8 alkenyl), -O-(C2-C8 alkynyl), -aryl, -C(O)R', -
OC(O)R', -
C(O)OR', -C(O)NH2,
-C(O)NHR', -C(O)N(R')2, -NHC(O)R', -SR', -SO3R', -S(O)2R', -S(O)R', -OH, =0, -
N3
-NH2, -NH(R'), -N(R')2 and -CN, where each R' is independently selected from -
H, -Ci-
C8 alkyl, -C2-C8 alkyenl, -C2-C8 alkynyl, or -aryl and wherein said -O-(C1-C8
alkyl), -0-
(C2-C8 alkenyl), -O-(C2-C8 alkynyl), -aryl, -CI-C8 alkyl, -C2-C8 alkenyl, and -
C2-C8
alkynyl groups can be optionally further substituted with one or more
substituents
including, but not limited to, -CI-C8 alkyl, -C2-C8 alkenyl, -C2-C8 alkynyl, -
halogen, -0-
(C1-C8 alkyl), -O-(C2-C8 alkenyl), -O-(C2C8 alkynyl), -aryl, -C(O)R", -
OC(O)R", -
C(O)OR", -C(O)NH2, -C(O)NHR", -C(O)N(R")2, -NHC(O)R", -SR",
-SO3R", -S(O)2R", -S(O)R", -OH, -N3, -NH2, -NH(R"), -N(R")2 and -CN, where
each
R" is independently selected from -H, -C1-C8 alkyl, -C2-C8 alkenyl, -C2-C8
alkynyl, or -
aryl.
[0069] Unless otherwise noted, the term "alkylene" refers to a saturated
branched or
straight chain hydrocarbon radical having from about 1 to about 20 carbon
atoms (and all
combinations and subcombinations of ranges and specific numbers of carbon
atoms
therein), with from about 1 to about 8 carbon atoms being preferred and having
two
monovalent radical centers derived by the removal of two hydrogen atoms from
the same
or two different carbon atoms of a parent alkane. Typical alkylenes include,
but are not
limited to, methylene, ethylene, propylene, butylene, pentylene, hexylene,
heptylene,
ocytylene, nonylene, decalene, 1,4-cyclohexylene, and the like. Alkylene
groups,
whether alone or as part of another group, can be optionally substituted with
one or more
groups, preferably 1 to 3 groups (and any additional substituents selected
from halogen),
including, but not limited to, -halogen, -O-(C1-C8 alkyl),

16


CA 02754531 2011-09-02
WO 2010/111018 PCT/US2010/026429
-O-(C2-C8 alkenyl), -O-(C2-C8 alkynyl), -aryl, -C(O)R', -OC(O)R', -C(O)OR', -
C(O)NH2,
-C(O)NHR', -C(O)N(R')2, -NHC(O)R', -SR', -SO3R', -S(O)2R', -S(O)R', -OH, =0, -
N3,
-NH2, -NH(R'), -N(R')2 and -CN, where each R' is independently selected from -
H, -Ci-
C8 alkyl, -C2-C8 alkenyl, -C2-C8 alkynyl, or -aryl and wherein said -O-(C1-C8
alkyl), -0-
(C2-C8 alkenyl), -O-(C2-C8 alkynyl), -aryl, -C1-C8 alkyl, -C2-C8 alkenyl, and -
C2-C8
alkynyl groups can be further optionally substituted with one or more
substituents
including, but not limited to, -C1-C8 alkyl, -C2-C8 alkenyl, -C2-C8 alkynyl, -
halogen, -0-
(C1-C8 alkyl), -O-(C2-C8 alkenyl), -O-(C2-C8 alkynyl), -aryl, -C(O)R", -
OC(O)R", -
C(O)OR", -C(O)NH2, -C(O)NHR", -C(O)N(R")2, -NHC(O)R", -SR", -SO3R", -
S(O)2R", -S(O)R", -OH, -N3, -NH2, -NH(R"), -N(R")2 and -CN, where each R" is
independently selected from -H, -C1-C8 alkyl, -C2-C8 alkenyl, -C2-C8 alkynyl,
or -aryl.
[0070] Unless otherwise noted, the term "alkenylene" refers to an optionally
substituted alkylene group containing at least one carbon-carbon double bond.
Exemplary alkenylene groups include, for example, ethenylene (-CH=CH-) and
propenylene (-CH=CHCH2-).
[0071] Unless otherwise noted, the term "alkynylene" refers to an optionally
substituted alkylene group containing at least one carbon-carbon triple bond.
Exemplary
alkynylene groups include, for example, acetylene (-C C-), propargyl (-CH2C=C-
), and
4-pentynyl (-CH2CH2CH2C CH-).
[0072] Unless otherwise noted, the term "aryl" refers to a monovalent aromatic
hydrocarbon radical of 6-20 carbon atoms (and all combinations and
subcombinations of
ranges and specific numbers of carbon atoms therein) derived by the removal of
one
hydrogen atom from a single carbon atom of a parent aromatic ring system. Some
aryl
groups are represented in the exemplary structures as "Ar". Typical aryl
groups include,
but are not limited to, radicals derived from benzene, substituted benzene,
phenyl,
naphthalene, anthracene, biphenyl, and the like.
[0073] An aryl group, whether alone or as part of another group, can be
optionally
substituted with one or more, preferably 1 to 5, or even 1 to 2 groups
including, but not
limited to, -halogen, -C1-C8 alkyl, -C2-C8 alkenyl, -C2-C8 alkynyl, -O-(C1-C8
alkyl), -0-
(C2-C8 alkenyl), -O-(C2-C8 alkynyl), -aryl, -C(O)R', -OC(O)R', -C(O)OR', -
C(O)NH2,
-C(O)NHR', -C(O)N(R')2,

17


CA 02754531 2011-09-02
WO 2010/111018 PCT/US2010/026429
-NHC(O)R', -SR', -SO3R', -S(O)2R', -S(O)R', -OH, -NO2, -N3, -NH2, -NH(R'),
-N(R')2 and -CN, where each R' is independently selected from -H, -C1-C8
alkyl, -C2-C8
alkenyl, -C2-C8 alkynyl, or -aryl and wherein said -C1-C8 alkyl, -C2-C8
alkenyl, -C2-C8
alkynyl, O-(C1-C8 alkyl), -O-(C2-C8 alkenyl), -O-(C2-C8 alkynyl), and -aryl
groups can
be further optionally substituted with one or more substituents including, but
not limited
to, -C1-C8 alkyl,
-C2-C8 alkenyl, -C2-C8 alkynyl, -halogen, -O-(C1-C8 alkyl), -O-(C2-C8
alkenyl), -O-(C2-C8
alkynyl), -aryl, -C(O)R", -OC(O)R", -C(O)OR", -C(O)NH2, -C(O)NHR", -
C(O)N(R" )2,
-NHC(O)R", -SR", -SO3R", -S(O)2R", -S(O)R", -OH, -N3 , -NH2, -NH(R"), -N(R")2
and
-CN, where each R" is independently selected from -H, -C1-C8 alkyl, -C2-C8
alkenyl, -C2-
C8 alkynyl, or -aryl.
[0074] Unless otherwise noted, the term "arylene" refers to an optionally
substituted aryl group which is divalent (i.e., derived by the removal of two
hydrogen
atoms from the same or two different carbon atoms of a parent aromatic ring
system) and
can be in the ortho, meta, or para configurations as shown in the following
structures with
phenyl as the exemplary aryl group.

j_rIIr
Typical "-(CI-C8 alkylene)aryl," "-(C2-C8 alkenylene)aryl", "and -(C2-C8
alkynylene)aryl" groups include, but are not limited to, benzyl, 2-phenylethan-
1-yl, 2-
phenylethen-1-yl, naphthylmethyl, 2-naphthylethan-1-yl, 2-naphthylethen-1-yl,
naphthobenzyl, 2-naphthophenylethan-1-yl and the like.

[0075] Unless otherwise noted, the term "heterocycle," refers to a monocyclic,
bicyclic, or polycyclic ring system having from 3 to 14 ring atoms (also
referred to as ring
members) wherein at least one ring atom in at least one ring is a heteroatom
selected from
N, 0, P, or S (and all combinations and subcombinations of ranges and specific
numbers
of carbon atoms and heteroatoms therein). The heterocycle can have from 1 to 4
ring
heteroatoms independently selected from N, 0, P, or S. One or more N, C, or S
atoms in

18


CA 02754531 2011-09-02
WO 2010/111018 PCT/US2010/026429
a heterocycle can be oxidized. A monocylic heterocycle preferably has 3 to 7
ring
members (e.g., 2 to 6 carbon atoms and 1 to 3 heteroatoms independently
selected from
N, 0, P, or S), and a bicyclic heterocycle preferably has 5 to 10 ring members
(e.g., 4 to
9 carbon atoms and 1 to 3 heteroatoms independently selected from N, 0, P, or
S). The
ring that includes the heteroatom can be aromatic or non-aromatic. Unless
otherwise
noted, the heterocycle is attached to its pendant group at any heteroatom or
carbon atom
that results in a stable structure.
[0076] Heterocycles are described in Paquette, "Principles of Modern
Heterocyclic
Chemistry" (W.A. Benjamin, New York, 1968), particularly Chapters 1, 3, 4, 6,
7, and 9;
The Chemistry of Heterocyclic Compounds, A series of Monographs" (John Wiley &
Sons, New York, 1950 to present), in particular Volumes 13, 14, 16, 19, and
28; and J.
Am. Chem. Soc. 82:5566 (1960).
[0077] Examples of "heterocycle" groups include by way of example and not
limitation pyridyl, dihydropyridyl, tetrahydropyridyl (piperidyl), thiazolyl,
pyrimidinyl,
furanyl, thienyl, pyrrolyl, pyrazolyl, imidazolyl, tetrazolyl, benzofuranyl,
thianaphthalenyl, indolyl, indolenyl, quinolinyl, isoquinolinyl,
benzimidazolyl,
piperidinyl, 4-piperidonyl, pyrrolidinyl, 2-pyrrolidonyl, pyrrolinyl,
tetrahydrofuranyl, bis-
tetrahydrofuranyl, tetrahydropyranyl, bis-tetrahydropyranyl,
tetrahydroquinolinyl,
tetrahydroisoquinolinyl, decahydroquinolinyl, octahydroisoquinolinyl,
azocinyl, triazinyl,
6H-1,2,5-thiadiazinyl, 2H,6H-1,5,2-dithiazinyl, thienyl, thianthrenyl,
pyranyl,
isobenzofuranyl, chromenyl, xanthenyl, phenoxathinyl, 2H-pyrrolyl,
isothiazolyl,
isoxazolyl, pyrazinyl, pyridazinyl, indolizinyl, isoindolyl, 3H-indolyl, 1H-
indazolyl,
purinyl, 4H-quinolizinyl, phthalazinyl, naphthyridinyl, quinoxalinyl,
quinazolinyl,
cinnolinyl, pteridinyl, 4H-carbazolyl, carbazolyl, 0-carbolinyl,
phenanthridinyl, acridinyl,
pyrimidinyl, phenanthrolinyl, phenazinyl, phenothiazinyl, furazanyl,
phenoxazinyl,
isochromanyl, chromanyl, imidazolidinyl, imidazolinyl, pyrazolidinyl,
pyrazolinyl,
piperazinyl, indolinyl, isoindolinyl, quinuclidinyl, morpholinyl,
oxazolidinyl,
benzotriazolyl, benzisoxazolyl, oxindolyl, benzoxazolinyl, and isatinoyl.
Preferred
"heterocycle" groups include, but are not limited to, benzofuranyl,
benzothiophenyl,
indolyl, benzopyrazolyl, coumarinyl, isoquinolinyl, pyrrolyl, thiophenyl,
furanyl,
thiazolyl, imidazolyl, pyrazolyl, triazolyl, quinolinyl, pyrimidinyl,
pyridinyl, pyridonyl,
pyrazinyl, pyridazinyl, isothiazolyl, isoxazolyl and tetrazolyl.

19


CA 02754531 2011-09-02
WO 2010/111018 PCT/US2010/026429
[0078] A heterocycle group, whether alone or as part of another group, can be
optionally substituted with one or more groups, preferably 1 to 2 groups,
including but
not limited to, -CI-C8 alkyl, -C2-C8 alkenyl, -C2-C8 alkynyl, -halogen, -O-(C1-
C8 alkyl), -
O-(C2-C8 alkenyl), -O-(C2-C8 alkynyl), -aryl, -C(O)R', -OC(O)R', -C(O)OR', -
C(O)NH2
, -C(O)NHR', -C(O)N(R')2,
-NHC(O)R', -SR', -SO3R', -S(O)2R', -S(O)R', -OH, -N3, -NH2, -NH(R'), -N(R')2
and
-CN, where each R' is independently selected from -H, -C1-C8 alkyl, -C2-C8
alkenyl, -C2-
C8 alkynyl, or -aryl and wherein said -O-(C1-C8 alkyl), -O-(C2-C8 alkenyl), -O-
(C2-C8
alkynyl), -C1-C8 alkyl, -C2-C8 alkenyl, -C2-C8 alkynyl, and -aryl groups can
be further
optionally substituted with one or more substituents including, but not
limited to, -C1-C8
alkyl, -C2-C8 alkenyl, -C2-C8 alkynyl,
-halogen, -O-(C1-C8 alkyl), -O-(C2-C8 alkenyl), -O-(C2-C8 alkynyl), -aryl, -
C(O)R", -
OC(O)R", -C(O)OR", -C(O)NH2, -C(O)NHR", -C(O)N(R")2, -NHC(O)R", -SR", -
SO3R", -S(O)2R",
-S(O)R", -OH, -N3, -NH2, -NH(R"), -N(R")2 and -CN, where each R" is
independently
selected from -H, -CI-C8 alkyl, -C2-C8 alkenyl, -C2-C8 alkynyl, or aryl.
[0079] By way of example and not limitation, carbon-bonded heterocycles can be
bonded at the following positions: position 2, 3, 4, 5, or 6 of a pyridine;
position 3, 4, 5,
or 6 of a pyridazine; position 2, 4, 5, or 6 of a pyrimidine; position 2, 3,
5, or 6 of a
pyrazine; position 2, 3, 4, or 5 of a furan, tetrahydrofuran, thiofuran,
thiophene, pyrrole or
tetrahydropyrrole; position 2, 4, or 5 of an oxazole, imidazole or thiazole;
position 3, 4, or
of an isoxazole, pyrazole, or isothiazole; position 2 or 3 of an aziridine;
position 2, 3, or
4 of an azetidine; position 2, 3, 4, 5, 6, 7, or 8 of a quinoline; or position
1, 3, 4, 5, 6, 7, or
8 of an isoquinoline. Still more typically, carbon bonded heterocycles include
2-pyridyl,
3-pyridyl, 4-pyridyl, 5-pyridyl, 6-pyridyl, 3-pyridazinyl, 4-pyridazinyl, 5-
pyridazinyl, 6-
pyridazinyl, 2-pyrimidinyl, 4-pyrimidinyl, 5-pyrimidinyl, 6-pyrimidinyl, 2-
pyrazinyl, 3-
pyrazinyl, 5-pyrazinyl, 6-pyrazinyl, 2-thiazolyl, 4-thiazolyl, or 5-thiazolyl.
[0080] By way of example and not limitation, nitrogen bonded heterocycles can
be
bonded at position 1 of an aziridine, azetidine, pyrrole, pyrrolidine, 2-
pyrroline, 3-
pyrroline, imidazole, imidazolidine, 2-imidazoline, 3-imidazoline, pyrazole,
pyrazoline,
2-pyrazoline, 3-pyrazoline, piperidine, piperazine, indole, indoline, or 1H-
indazole;
position 2 of a isoindole, or isoindoline; position 4 of a morpholine; and
position 9 of a



CA 02754531 2011-09-02
WO 2010/111018 PCT/US2010/026429
carbazole, or (3-carboline. Still more typically, nitrogen bonded heterocycles
include 1-
aziridyl, 1-azetedyl, 1-pyrrolyl, 1-imidazolyl, 1-pyrazolyl, and 1-
piperidinyl.
[0081] Unless otherwise noted, the term "carbocycle," refers to a saturated or
unsaturated non-aromatic monocyclic, bicyclic, or polycyclic ring system
having from 3
to 14 ring atoms (and all combinations and subcombinations of ranges and
specific
numbers of carbon atoms therein) wherein all of the ring atoms are carbon
atoms.
Monocyclic carbocycles preferably have 3 to 6 ring atoms, still more
preferably 5 or 6
ring atoms. Bicyclic carbocycles preferably have 7 to 12 ring atoms, e.g.,
arranged as a
bicyclo [4,5], [5,5], [5,6] or [6,6] system, or 9 or 10 ring atoms arranged as
a bicyclo [5,6]
or [6,6] system. The term "carbocycle" includes, for example, a monocyclic
carbocycle
ring fused to an aryl ring (e.g., a monocyclic carbocycle ring fused to a
benzene ring).
Carbocyles preferably have 3 to 8 carbon ring atoms.
[0082] Carbocycle groups, whether alone or as part of another group, can be
optionally substituted with, for example, one or more groups, preferably 1 or
2 groups
(and any additional substituents selected from halogen), including, but not
limited to, -
halogen, -C1-C8 alkyl, -C2-C8 alkenyl, -C2-C8 alkynyl, -0-(C1-C8 alkyl), -O-
(C2-C8
alkenyl), -0-(C2-C8 alkynyl), -aryl,
-C(O)R', -OC(O)R', -C(O)OR', -C(O)NH2 , -C(O)NHR', -C(O)N(R')2, -NHC(O)R', -
SR',
-S03R', -S(0)2R', -S(O)R', -OH, =0, -N3, -NH2, -NH(R'), -N(R')2 and -CN, where
each
R' is independently selected from -H, -CI-C8 alkyl, -C2-C8 alkenyl, -C2-C8
alkynyl, or -
aryl and wherein said -C1-C8 alkyl, -C2-C8 alkenyl, -C2-C8 alkynyl, -0-(C1-C8
alkyl), -0-
(C2-C8 alkenyl), -0-(C2-C8 alkynyl), and -aryl groups can be further
optionally
substituted with one or more substituents including, but not limited to, -C1-
C8 alkyl, -C2-
C8 alkenyl, -C2-C8 alkynyl, -halogen, -0-(C1-C8 alkyl), -0-(C2-C8 alkenyl), -0-
(C2-C8
alkynyl), -aryl, -C(O)R", -OC(O)R",
-C(O)OR", -C(O)NH2 , -C(O)NHR", -C(O)N(R")2, -NHC(O)R", -SR", -S03R", -
S(0)2R",
-S(O)R", -OH, -N3, -NH2, -NH(R"), -N(R")2 and -CN, where each R" is
independently selected from -H, -C1-C8 alkyl, -C2-C8 alkenyl, -C2-C8 alkynyl,
or -aryl.
[0083] Examples of monocyclic carbocylic substituents include -cyclopropyl,
-cyclobutyl, -cyclopentyl, -1-cyclopent-l-enyl, -1-cyclopent-2-enyl, -1-
cyclopent-3-enyl,
21


CA 02754531 2011-09-02
WO 2010/111018 PCT/US2010/026429
cyclohexyl, -1-cyclohex-l-enyl, -1-cyclohex-2-enyl, -1-cyclohex-3-enyl, -
cycloheptyl,
-cyclooctyl. -1,3-cyclohexadienyl, -1,4-cyclohexadienyl, -1,3-
cycloheptadienyl,
-1,3,5-cycloheptatrienyl, and -cyclooctadienyl.
[0084] A "carbocyclo," whether used alone or as part of another group, refers
to an
optionally substituted carbocycle group as defined above that is divalent
(i.e., derived by
the removal of two hydrogen atoms from the same or two different carbon atoms
of a
parent carbocyclic ring system).
[0085] Unless otherwise indicated by context, a hyphen (-) designates the
point of
attachment to the pendant molecule. Accordingly, the term "-(C1-C8
alkylene)aryl" or
"-Ci-C8 alkylene(aryl)" refers to a CI-C8 alkylene radical as defined herein
wherein the
alkylene radical is attached to the pendant molecule at any of the carbon
atoms of the
alkylene radical and one of the hydrogen atoms bonded to a carbon atom of the
alkylene
radical is replaced with an aryl radical as defined herein.
[0086] When a particular group is "substituted", that group may have one or
more
substituents, preferably from one to five substituents, more preferably from
one to three
substituents, most preferably from one to two substituents, independently
selected from
the list of substituents. The group can, however, generally have any number of
substituents selected from halogen. Groups that are substituted are so
indicated.
[0087] It is intended that the definition of any substituent or variable at a
particular
location in a molecule be independent of its definitions elsewhere in that
molecule. It is
understood that substituents and substitution patterns on the compounds of
this invention
can be selected by one of ordinary skill in the art to provide compounds that
are
chemically stable and that can be readily synthesized by techniques known in
the art as
well as those methods set forth herein.
[0088] Protective groups as used herein refer to groups which selectively
block, either
temporarily or permanently, one reactive site in a multifunctional compound.
Suitable
hydroxy-protecting groups for use in the present invention are
pharmaceutically
acceptable and may or may not need to be cleaved from the parent compound
after
administration to a subject in order for the compound to be active. Cleavage
is through
normal metabolic processes within the body. Hydroxy protecting groups are well
known
in the art, see, Protective Groups in Organic Synthesis by T. W. Greene and P.
G. M.
Wuts (John Wiley & sons, 3rd Edition) incorporated herein by reference in its
entirety and

22


CA 02754531 2011-09-02
WO 2010/111018 PCT/US2010/026429
for all purposes and include, for example, ether (e.g., alkyl ethers and silyl
ethers
including, for example, dialkylsilylether, trialkylsilylether,
dialkylalkoxysilylether), ester,
carbonate, carbamates, sulfonate, and phosphate protecting groups. Examples of
hydroxy
protecting groups include, but are not limited to, methyl ether; methoxymethyl
ether,
methylthiomethyl ether, (phenyldimethylsilyl)methoxymethyl ether,
benzyloxymethyl
ether, p-methoxybenzyloxymethyl ether, p-nitrobenzyloxymethyl ether, o-
nitrobenzyloxymethyl ether, (4-methoxyphenoxy)methyl ether, guaiacolmethyl
ether, t-
butoxymethyl ether, 4-pentenyloxymethyl ether, siloxymethyl ether, 2-
methoxyethoxymethyl ether, 2,2,2-trichloroethoxymethyl ether, bis(2-
chloroethoxy)methyl ether, 2-(trimethylsilyl)ethoxymethyl ether,
menthoxymethyl ether,
tetrahydropyranyl ether, 1-methoxycylcohexyl ether, 4-
methoxytetrahydrothiopyranyl
ether, 4-methoxytetrahydrothiopyranyl ether S,S-Dioxide, 1-[(2-choro-4-
methyl)phenyl]-
4-methoxypiperidin-4-yl ether, 1-(2-fluorophneyl)-4-methoxypiperidin-4-yl
ether, 1,4-
dioxan-2-yl ether, tetrahydrofuranyl ether, tetrahydrothiofuranyl ether;
substituted ethyl
ethers such as 1-ethoxyethyl ether, 1-(2-chloroethoxy)ethyl ether, 1-[2-
(trimethylsilyl)ethoxy] ethyl ether, 1-methyl-l-methoxyethyl ether, 1-methyl-l-

benzyloxyethyl ether, 1-methyl-l-benzyloxy-2-fluoroethyl ether, 1-methyl-
lphenoxyethyl
ether, 2-trimethylsilyl ether, t-butyl ether, allyl ether, propargyl ethers, p-
chlorophenyl
ether, p-methoxyphenyl ether, benzyl ether, p-methoxybenzyl ether 3,4-
dimethoxybenzyl
ether, trimethylsilyl ether, triethylsilyl ether, tripropylsilylether,
dimethylisopropylsilyl
ether, diethylisopropylsilyl ether, dimethylhexylsilyl ether, t-
butyldimethylsilyl ether,
diphenylmethylsilyl ether, benzoylformate ester, acetate ester, chloroacetate
ester,
dichloroacetate ester, trichloroacetate ester, trifluoroacetate ester,
methoxyacetate ester,
triphneylmethoxyacetate ester, phenylacetate ester, benzoate ester, alkyl
methyl
carbonate, alkyl 9-fluorenylmethyl carbonate, alkyl ethyl carbonate, alkyl
2,2,2,-
trichloroethyl carbonate, 1,1,-dimethyl-2,2,2-trichloroethyl carbonate,
alkylsulfonate,
methanesulfonate, benzylsulfonate, tosylate, methylene acetal, ethylidene
acetal, and t-
butylmethylidene ketal. Preferred protecting groups are represented by the
formulas -Ra,
-Si(Ra)(Ra)(Ra), -C(O)Ra, -C(O)OR a, -C(O)NH(Ra), -S(O)2Ra, -S(O)20H,
P(O)(OH)2, and
-P(O)(OH)ORa, wherein Ra is CI-C20 alkyl, C2-C20 alkenyl, C2-C20 alkynyl, -CI-
C20
alkylene(carbocycle), -C2-C20 alkenylene(carbocycle), -C2-C20
alkynylene(carbocycle), -
C6-CIO aryl, -CI-C20 alkylene(aryl), -C2-C20 alkenylene(aryl), -C2-C20
alkynylene(aryl), -

23


CA 02754531 2011-09-02
WO 2010/111018 PCT/US2010/026429
CI-C20 alkylene(heterocycle), -C2-C20 alkenylene(heterocycle), or -C2-C20
alkynylene(heterocycle) wherein said alkyl, alkenyl, alkynyl, alkylene,
alkenylene,
alkynylene, aryl, carbocycle, and heterocycle radicals whether alone or as
part of another
group are optionally substituted.
[0089] "Altering the native glycosylation pattern" is intended for purposes
herein to
mean deleting one or more carbohydrate moieties found in native sequence
24P4C12
(either by removing the underlying glycosylation site or by deleting the
glycosylation by
chemical and/or enzymatic means), and/or adding one or more glycosylation
sites that are
not present in the native sequence 24P4C12. In addition, the phrase includes
qualitative
changes in the glycosylation of the native proteins, involving a change in the
nature and
proportions of the various carbohydrate moieties present.
[0090] The term "analog" refers to a molecule which is structurally similar or
shares
similar or corresponding attributes with another molecule (e.g. a 24P4C 12-
related
protein). For example, an analog of a 24P4C12 protein can be specifically
bound by an
antibody or T cell that specifically binds to 24P4C12.
[0091] The term "antibody" is used in the broadest sense unless clearly
indicated
otherwise. Therefore, an "antibody" can be naturally occurring or man-made
such as
monoclonal antibodies produced by conventional hybridoma technology. 24P4C12
antibodies comprise monoclonal and polyclonal antibodies as well as fragments
containing the antigen-binding domain and/or one or more complementarity
determining
regions of these antibodies. As used herein, the term "antibody" refers to any
form of
antibody or fragment thereof that specifically binds 24P4C12 and/or exhibits
the desired
biological activity and specifically covers monoclonal antibodies (including
full length
monoclonal antibodies), polyclonal antibodies, multispecific antibodies (e.g.,
bispecific
antibodies), and antibody fragments so long as they specifically bind 24P4C12
and/or
exhibit the desired biological activity. Any specific antibody can be used in
the methods
and compositions provided herein. Thus, in one embodiment the term "antibody"
encompasses a molecule comprising at least one variable region from a light
chain
immunoglobulin molecule and at least one variable region from a heavy chain
molecule
that in combination form a specific binding site for the target antigen. In
one
embodiment, the antibody is an IgG antibody. For example, the antibody is a
IgGi, IgG2,
IgG3, or IgG4 antibody. The antibodies useful in the present methods and
compositions

24


CA 02754531 2011-09-02
WO 2010/111018 PCT/US2010/026429
can be generated in cell culture, in phage, or in various animals, including
but not limited
to cows, rabbits, goats, mice, rats, hamsters, guinea pigs, sheep, dogs, cats,
monkeys,
chimpanzees, and apes. Therefore, in one embodiment, an antibody of the
present
invention is a mammalian antibody. Phage techniques can be used to isolate an
initial
antibody or to generate variants with altered specificity or avidity
characteristics. Such
techniques are routine and well known in the art. In one embodiment, the
antibody is
produced by recombinant means known in the art. For example, a recombinant
antibody
can be produced by transfecting a host cell with a vector comprising a DNA
sequence
encoding the antibody. One or more vectors can be used to transfect the DNA
sequence
expressing at least one VL and one VH region in the host cell. Exemplary
descriptions of
recombinant means of antibody generation and production include Delves,
ANTIBODY
PRODUCTION: ESSENTIAL TECHNIQUES (Wiley, 1997); Shephard, et al.,
MONOCLONAL ANTIBODIES (Oxford University Press, 2000); Goding,
MONOCLONAL ANTIBODIES: PRINCIPLES AND PRACTICE (Academic Press,
1993); and CURRENT PROTOCOLS IN IMMUNOLOGY (John Wiley & Sons, most
recent edition). An antibody of the present invention can be modified by
recombinant
means to increase efficacy of the antibody in mediating the desired function.
Thus, it is
within the scope of the invention that antibodies can be modified by
substitutions using
recombinant means. Typically, the substitutions will be conservative
substitutions. For
example, at least one amino acid in the constant region of the antibody can be
replaced
with a different residue. See, e.g., U.S. Patent No. 5,624,821, U.S. Patent
No. 6,194,551,
Application No. WO 9958572; and Angal, et al., Mol. Immunol. 30: 105-08
(1993). The
modification in amino acids includes deletions, additions, and substitutions
of amino
acids. In some cases, such changes are made to reduce undesired activities,
e.g.,
complement-dependent cytotoxicity. Frequently, the antibodies are labeled by
joining,
either covalently or non-covalently, a substance which provides for a
detectable signal. A
wide variety of labels and conjugation techniques are known and are reported
extensively
in both the scientific and patent literature. These antibodies can be screened
for binding
to normal or defective 24P4C12. See e.g., ANTIBODY ENGINEERING: A
PRACTICAL APPROACH (Oxford University Press, 1996). Suitable antibodies with
the desired biologic activities can be identified using the following in vitro
assays
including but not limited to: proliferation, migration, adhesion, soft agar
growth,



CA 02754531 2011-09-02
WO 2010/111018 PCT/US2010/026429
angiogenesis, cell-cell communication, apoptosis, transport, signal
transduction, and the
following in vivo assays such as the inhibition of tumor growth. The
antibodies provided
herein can also be useful in diagnostic applications. As capture or non-
neutralizing
antibodies, they can be screened for the ability to bind to the specific
antigen without
inhibiting the receptor-binding or biological activity of the antigen. As
neutralizing
antibodies, the antibodies can be useful in competitive binding assays. They
can also be
used to quantify the 24P4C12 or its receptor.
[0092] The term "antigen-binding portion" or "antibody fragment" of an
antibody (or
simply "antibody portion"), as used herein, refers to one or more fragments of
a 24P4C12
antibody that retain the ability to specifically bind to an antigen (e.g.,
24P4C12 and
variants; Figure 1). It has been shown that the antigen-binding function of an
antibody
can be performed by fragments of a full-length antibody. Examples of binding
fragments
encompassed within the term "antigen-binding portion" of an antibody include
(i) a Fab
fragment, a monovalent fragment consisting of the VL, VH, CL and CHI domains;
(ii) a
F(ab')2 fragment, a bivalent fragment comprising two Fab fragments linked by a
disulfide
bridge at the hinge region; (iii) a Fd fragment consisting of the VH and CHI
domains; (iv)
a Fv fragment consisting of the VL and VH domains of a single arm of an
antibody, (v) a
dAb fragment (Ward et al., (1989) Nature 341:544-546), which consists of a VH
domain;
and (vi) an isolated complementarily determining region (CDR). Furthermore,
although
the two domains of the Fv fragment, VL and VH, are coded for by separate
genes, they can
be joined, using recombinant methods, by a synthetic linker that enables them
to be made
as a single protein chain in which the VL and VH regions pair to form
monovalent
molecules (known as single chain Fv (scFv); see e.g., Bird et al. (1988)
Science 242:423-
426; and Huston et al. (1988) Proc. Natl. Acad. Sci. USA 85:5879-5883). Such
single
chain antibodies are also intended to be encompassed within the term "antigen-
binding
portion" of an antibody. These antibody fragments are obtained using
conventional
techniques known to those with skill in the art, and the fragments are
screened for utility
in the same manner as are intact antibodies.
[0093] As used herein, any form of the "antigen" can be used to generate an
antibody
that is specific for 24P4C 12. Thus, the eliciting antigen may be a single
epitope, multiple
epitopes, or the entire protein alone or in combination with one or more
immunogenicity
enhancing agents known in the art. The eliciting antigen may be an isolated
full-length

26


CA 02754531 2011-09-02
WO 2010/111018 PCT/US2010/026429
protein, a cell surface protein (e.g., immunizing with cells transfected with
at least a
portion of the antigen), or a soluble protein (e.g., immunizing with only the
extracellular
domain portion of the protein). The antigen may be produced in a genetically
modified
cell. The DNA encoding the antigen may be genomic or non-genomic (e.g., cDNA)
and
encodes at least a portion of the extracellular domain. As used herein, the
term "portion"
refers to the minimal number of amino acids or nucleic acids, as appropriate,
to constitute
an immunogenic epitope of the antigen of interest. Any genetic vectors
suitable for
transformation of the cells of interest may be employed, including but not
limited to
adenoviral vectors, plasmids, and non-viral vectors, such as cationic lipids.
In one
embodiment, the antibody of the methods and compositions herein specifically
bind at
least a portion of the extracellular domain of the 24P4C 12 of interest.
[0094] The antibodies or antigen binding fragments thereof provided herein may
be
conjugated to a "bioactive agent." As used herein, the term "bioactive agent"
refers to
any synthetic or naturally occurring compound that binds the antigen and/or
enhances or
mediates a desired biological effect to enhance cell-killing toxins. In one
embodiment,
the binding fragments useful in the present invention are biologically active
fragments.
As used herein, the term "biologically active" refers to an antibody or
antibody fragment
that is capable of binding the desired antigenic epitope and directly or
indirectly exerting
a biologic effect. Direct effects include, but are not limited to the
modulation,
stimulation, and/ or inhibition of a growth signal, the modulation,
stimulation, and/ or
inhibition of an anti-apoptotic signal, the modulation, stimulation, and/ or
inhibition of an
apoptotic or necrotic signal, modulation, stimulation, and/ or inhibition the
ADCC
cascade, and modulation, stimulation, and/ or inhibition the CDC cascade.
[0095] "Bispecific" antibodies are also useful in the present methods and
compositions. As used herein, the term "bispecific antibody" refers to an
antibody,
typically a monoclonal antibody, having binding specificities for at least two
different
antigenic epitopes. In one embodiment, the epitopes are from the same antigen.
In
another embodiment, the epitopes are from two different antigens. Methods for
making
bispecific antibodies are known in the art. For example, bispecific antibodies
can be
produced recombinantly using the co-expression of two immunoglobulin heavy
chain/light chain pairs. See, e.g., Milstein et al., Nature 305:537-39 (1983).
Alternatively, bispecific antibodies can be prepared using chemical linkage.
See, e.g.,

27


CA 02754531 2011-09-02
WO 2010/111018 PCT/US2010/026429
Brennan, et al., Science 229:81 (1985). Bispecific antibodies include
bispecific antibody
fragments. See, e.g., Hollinger, et al., Proc. Natl. Acad. Sci. U.S.A. 90:6444-
48 (1993),
Gruber, et al., J. Immunol. 152:5368 (1994).
[0096] The monoclonal antibodies described herein specifically include
"chimeric"
antibodies in which a portion of the heavy and/or light chain is identical
with or
homologous to corresponding sequences in antibodies derived from a particular
species or
belonging to a particular antibody class or subclass, while the remainder of
the chain(s) is
identical with or homologous to corresponding sequences in antibodies derived
from
another species or belonging to another antibody class or subclass, as well as
fragments of
such antibodies, so long as they specifically bind the target antigen and/or
exhibit the
desired biological activity (U.S. Pat. No. 4,816,567; and Morrison et al.,
Proc. Natl. Acad.
Sci. USA 81: 6851-6855 (1984)).
[0097] The term "Chemotherapeutic Agent" refers to all chemical compounds that
are
effective in inhibiting tumor growth. Non-limiting examples of
chemotherapeutic agents
include alkylating agents; for example, nitrogen mustards, ethyleneimine
compounds and
alkyl sulphonates; antimetabolites, for example, folic acid, purine or
pyrimidine
antagonists; mitotic inhibitors, for example, anti-tubulin agents such as
vinca alkaloids,
auristatins and derivatives of podophyllotoxin; cytotoxic antibiotics;
compounds that
damage or interfere with DNA expression or replication, for example, DNA minor
groove
binders; and growth factor receptor antagonists. In addition, chemotherapeutic
agents
include cytotoxic agents (as defined herein), antibodies, biological molecules
and small
molecules.
[0098] The term "compound" refers to and encompasses the chemical compound
itself as well as, whether explicitly stated or not, and unless the context
makes clear that
the following are to be excluded: amorphous and crystalline forms of the
compound,
including polymorphic forms, where these forms may be part of a mixture or in
isolation;
free acid and free base forms of the compound, which are typically the forms
shown in
the structures provided herein; isomers of the compound, which refers to
optical isomers,
and tautomeric isomers, where optical isomers include enantiomers and
diastereomers,
chiral isomers and non-chiral isomers, and the optical isomers include
isolated optical
isomers as well as mixtures of optical isomers including racemic and non-
racemic
mixtures; where an isomer may be in isolated form or in a mixture with one or
more other

28


CA 02754531 2011-09-02
WO 2010/111018 PCT/US2010/026429
isomers; isotopes of the compound, including deuterium- and tritium-containing
compounds, and including compounds containing radioisotopes, including
therapeutically- and diagnostically-effective radioisotopes; multimeric forms
of the
compound, including dimeric, trimeric, etc. forms; salts of the compound,
preferably
pharmaceutically acceptable salts, including acid addition salts and base
addition salts,
including salts having organic counterions and inorganic counterions, and
including
zwitterionic forms, where if a compound is associated with two or more
counterions, the
two or more counterions may be the same or different; and solvates of the
compound,
including hemisolvates, monosolvates, disolvates, etc., including organic
solvates and
inorganic solvates, said inorganic solvates including hydrates; where if a
compound is
associated with two or more solvent molecules, the two or more solvent
molecules may
be the same or different. In some instances, reference made herein to a
compound of the
invention will include an explicit reference to one or of the above forms,
e.g., salts and/or
solvates; however, this reference is for emphasis only, and is not to be
construed as
excluding other of the above forms as identified above.
[0099] As used herein, the term "conservative substitution" refers to
substitutions of
amino acids are known to those of skill in this art and may be made generally
without
altering the biological activity of the resulting molecule. Those of skill in
this art
recognize that, in general, single amino acid substitutions in non-essential
regions of a
polypeptide do not substantially alter biological activity (see, e.g., Watson,
et al.,
MOLECULAR BIOLOGY OF THE GENE, The Benjamin/Cummings Pub. Co., p. 224
(4th Edition 1987)). Such exemplary substitutions are preferably made in
accordance
with those set forth in Table II and Table(s) III(a-b). For example, such
changes include
substituting any of isoleucine (I), valine (V), and leucine (L) for any other
of these
hydrophobic amino acids; aspartic acid (D) for glutamic acid (E) and vice
versa;
glutamine (Q) for asparagine (N) and vice versa; and serine (S) for threonine
(T) and vice
versa. Other substitutions can also be considered conservative, depending on
the
environment of the particular amino acid and its role in the three-dimensional
structure of
the protein. For example, glycine (G) and alanine (A) can frequently be
interchangeable,
as can alanine (A) and valine (V). Methionine (M), which is relatively
hydrophobic, can
frequently be interchanged with leucine and isoleucine, and sometimes with
valine.
Lysine (K) and arginine (R) are frequently interchangeable in locations in
which the

29


CA 02754531 2011-09-02
WO 2010/111018 PCT/US2010/026429
significant feature of the amino acid residue is its charge and the differing
pK's of these
two amino acid residues are not significant. Still other changes can be
considered
"conservative" in particular environments (see, e.g. Table 111(a) herein;
pages 13-15
"Biochemistry" 2nd ED. Lubert Stryer ed (Stanford University); Henikoff et
al., PNAS
1992 Vol 89 10915-10919; Lei et al., J Biol Chem 1995 May 19; 270(20):11882-
6).
Other substitutions are also permissible and may be determined empirically or
in accord
with known conservative substitutions.
[00100] The term "cytotoxic agent" refers to a substance that inhibits or
prevents the
expression activity of cells, function of cells and/or causes destruction of
cells. The term
is intended to include radioactive isotopes, chemotherapeutic agents, and
toxins such as
small molecule toxins or enzymatically active toxins of bacterial, fungal,
plant or animal
origin, including fragments and/or variants thereof. Examples of cytotoxic
agents
include, but are not limited to auristatins (e.g., auristatin E, auristatin F,
MMAE and
MMAF), auromycins, maytansinoids, ricin, ricin A-chain, combrestatin,
duocarmycins,
dolastatins, doxorubicin, daunorubicin, taxols, cisplatin, cc 1065, ethidium
bromide,
mitomycin, etoposide, tenoposide, vincristine, vinblastine, colchicine,
dihydroxy
anthracin dione, actinomycin, diphtheria toxin, Pseudomonas exotoxin (PE) A,
PE40,
abrin, abrin A chain, modeccin A chain, alpha-sarcin, gelonin, mitogellin,
retstrictocin,
phenomycin, enomycin, curicin, crotin, calicheamicin, Sapaonaria officinalis
inhibitor,
and glucocorticoid and other chemotherapeutic agents, as well as radioisotopes
such as

At211, 1131, 1 125, Y9o Re186, Re'88, Sm153 Bi212 or 213, p32 and radioactive
isotopes of Lu

including Lu'77. Antibodies may also be conjugated to an anti-cancer pro-drug
activating
enzyme capable of converting the pro-drug to its active form.
[00101] As used herein, the term "dabodies" refers to small antibody fragments
with
two antigen-binding sites, which fragments comprise a heavy chain variable
domain (VH)
connected to a light chain variable domain (VL) in the same polypeptide chain
(VH-VL).
By using a linker that is too short to allow pairing between the two domains
on the same
chain, the domains are forced to pair with the complementary domains of
another chain
and create two antigen-binding sites. Diabodies are described more fully in,
e.g., EP
404,097; WO 93/11161; and Hollinger et al., Proc. Natl. Acad. Sci. USA 90:6444-
48
(1993).



CA 02754531 2011-09-02
WO 2010/111018 PCT/US2010/026429
[00102] The term "deplete," in the context of the effect of a 24P4C12 binding
agent on
24P4C 12-expres sing cells, refers to a reduction in the number of or
elimination of the
24P4C12-expressing cells.
[00103] The term "gene product" is used herein to indicate a peptide/protein
or
mRNA. For example, a "gene product of the invention" is sometimes referred to
herein
as a "cancer amino acid sequence", "cancer protein", "protein of a cancer
listed in Table
I", a "cancer mRNA", "mRNA of a cancer listed in Table I", etc. In one
embodiment, the
cancer protein is encoded by a nucleic acid of Figure 1. The cancer protein
can be a
fragment, or alternatively, be the full-length protein encoded by nucleic
acids of Figure 1.
In one embodiment, a cancer amino acid sequence is used to determine sequence
identity
or similarity. In another embodiment, the sequences are naturally occurring
allelic
variants of a protein encoded by a nucleic acid of Figure 1. In another
embodiment, the
sequences are sequence variants as further described herein.
[00104] "Heteroconjugate" antibodies are useful in the present methods and
compositions. As used herein, the term "heteroconjugate antibody" refers to
two
covalently joined antibodies. Such antibodies can be prepared using known
methods in
synthetic protein chemistry, including using crosslinking agents. See, e.g.,
U.S. Patent
No. 4,676,980.
[00105] The term "homolog" refers to a molecule which exhibits homology to
another
molecule, by for example, having sequences of chemical residues that are the
same or
similar at corresponding positions.
[00106] In one embodiment, the antibody provided herein is a "human antibody."
As
used herein, the term "human antibody" refers to an antibody in which
essentially the
entire sequences of the light chain and heavy chain sequences, including the
complementary determining regions (CDRs), are from human genes. In one
embodiment,
human monoclonal antibodies are prepared by the trioma technique, the human B-
cell
technique (see, e.g., Kozbor, et al., Immunol. Today 4: 72 (1983), EBV
transformation
technique (see, e.g., Cole et al. MONOCLONAL ANTIBODIES AND CANCER
THERAPY 77-96 (1985)), or using phage display (see, e.g., Marks et al., J.
Mol. Biol.
222:581 (1991)). In a specific embodiment, the human antibody is generated in
a
transgenic mouse. Techniques for making such partially to fully human
antibodies are
known in the art and any such techniques can be used. According to one
particularly

31


CA 02754531 2011-09-02
WO 2010/111018 PCT/US2010/026429
preferred embodiment, fully human antibody sequences are made in a transgenic
mouse
engineered to express human heavy and light chain antibody genes. An exemplary
description of preparing transgenic mice that produce human antibodies found
in
Application No. WO 02/43478 and United States Patent 6,657,103 (Abgenix) and
its
progeny. B cells from transgenic mice that produce the desired antibody can
then be
fused to make hybridoma cell lines for continuous production of the antibody.
See, e.g.,
U.S. Patent Nos. 5,569,825; 5,625,126; 5,633,425; 5,661,016; and 5,545,806;
and
Jakobovits, Adv. Drug Del. Rev. 31:33-42 (1998); Green, et al., J. Exp. Med.
188:483-95
(1998).
[00107] As used herein, the term "humanized antibody" refers to forms of
antibodies
that contain sequences from non-human (e.g., murine) antibodies as well as
human
antibodies. Such antibodies are chimeric antibodies which contain minimal
sequence
derived from non-human immunoglobulin. In general, the humanized antibody will
comprise substantially all of at least one, and typically two, variable
domains, in which all
or substantially all of the hypervariable loops correspond to those of a non-
human
immunoglobulin and all or substantially all of the FR regions are those of a
human
immunoglobulin sequence. The humanized antibody optionally also will comprise
at
least a portion of an immunoglobulin constant region (Fc), typically that of a
human
immunoglobulin. See e.g., Cabilly U.S. Patent No. 4,816,567; Queen et al.
(1989) Proc.
Nat'l Acad. Sci. USA 86:10029-10033; and ANTIBODY ENGINEERING: A
PRACTICAL APPROACH (Oxford University Press 1996).
[00108] The terms "inhibit" or "inhibition of' as used herein means to reduce
by a
measurable amount, or to prevent entirely.
[00109] The phrases "isolated" or "biologically pure" refer to material which
is
substantially or essentially free from components which normally accompany the
material
as it is found in its native state. Thus, isolated peptides in accordance with
the invention
preferably do not contain materials normally associated with the peptides in
their in situ
environment. For example, a polynucleotide is said to be "isolated" when it is
substantially separated from contaminant polynucleotides that correspond or
are
complementary to genes other than the 24P4C 12 genes or that encode
polypeptides other
than 24P4C12 gene product or fragments thereof. A skilled artisan can readily
employ
nucleic acid isolation procedures to obtain an isolated 24P4C12
polynucleotide. A

32


CA 02754531 2011-09-02
WO 2010/111018 PCT/US2010/026429
protein is said to be "isolated," for example, when physical, mechanical or
chemical
methods are employed to remove the 24P4C 12 proteins from cellular
constituents that are
normally associated with the protein. A skilled artisan can readily employ
standard
purification methods to obtain an isolated 24P4C12 protein. Alternatively, an
isolated
protein can be prepared by chemical means.
[00110] Suitable "labels" include radionuclides, enzymes, substrates,
cofactors,
inhibitors, fluorescent moieties, chemiluminescent moieties, magnetic
particles, and the
like. Patents teaching the use of such labels include U.S. Patent Nos.
3,817,837;
3,850,752; 3,939,350; 3,996,345; 4,277,437; 4,275,149; and 4,366,241. In
addition, the
antibodies provided herein can be useful as the antigen-binding component of
fluorobodies. See e.g., Zeytun et al., Nat. Biotechnol. 21:1473-79 (2003).
[00111] The term "mammal" refers to any organism classified as a mammal,
including
mice, rats, rabbits, dogs, cats, cows, horses and humans. In one embodiment of
the
invention, the mammal is a mouse. In another embodiment of the invention, the
mammal
is a human.
[00112] The terms "metastatic cancer" and "metastatic disease" mean cancers
that
have spread to regional lymph nodes or to distant sites, and are meant to
include stage D
disease under the AUA system and stage TxNxM+ under the TNM system.
[00113] The term "modulator" or "test compound" or "drug candidate" or
grammatical
equivalents as used herein describe any molecule, e.g., protein, oligopeptide,
small
organic molecule, polysaccharide, polynucleotide, etc., to be tested for the
capacity to
directly or indirectly alter the cancer phenotype or the expression of a
cancer sequence,
e.g., a nucleic acid or protein sequences, or effects of cancer sequences
(e.g., signaling,
gene expression, protein interaction, etc.) In one aspect, a modulator will
neutralize the
effect of a cancer protein of the invention. By "neutralize" is meant that an
activity of a
protein is inhibited or blocked, along with the consequent effect on the cell.
In another
aspect, a modulator will neutralize the effect of a gene, and its
corresponding protein, of
the invention by normalizing levels of said protein. In preferred embodiments,
modulators alter expression profiles, or expression profile nucleic acids or
proteins
provided herein, or downstream effector pathways. In one embodiment, the
modulator
suppresses a cancer phenotype, e.g. to a normal tissue fingerprint. In another
embodiment, a modulator induced a cancer phenotype. Generally, a plurality of
assay

33


CA 02754531 2011-09-02
WO 2010/111018 PCT/US2010/026429
mixtures is run in parallel with different agent concentrations to obtain a
differential
response to the various concentrations. Typically, one of these concentrations
serves as a
negative control, i.e., at zero concentration or below the level of detection.
[00114] Modulators, drug candidates, or test compounds encompass numerous
chemical classes, though typically they are organic molecules, preferably
small organic
compounds having a molecular weight of more than 100 and less than about 2,500
Daltons. Preferred small molecules are less than 2000, or less than 1500 or
less than 1000
or less than 500 D. Candidate agents comprise functional groups necessary for
structural
interaction with proteins, particularly hydrogen bonding, and typically
include at least an
amine, carbonyl, hydroxyl or carboxyl group, preferably at least two of the
functional
chemical groups. The candidate agents often comprise cyclical carbon or
heterocyclic
structures and/or aromatic or polyaromatic structures substituted with one or
more of the
above functional groups. Modulators also comprise biomolecules such as
peptides,
saccharides, fatty acids, steroids, purines, pyrimidines, derivatives,
structural analogs or
combinations thereof. Particularly preferred are peptides. One class of
modulators are
peptides, for example of from about five to about 35 amino acids, with from
about five to
about 20 amino acids being preferred, and from about 7 to about 15 being
particularly
preferred. Preferably, the cancer modulatory protein is soluble, includes a
non-
transmembrane region, and/or, has an N-terminal Cys to aid in solubility. In
one
embodiment, the C-terminus of the fragment is kept as a free acid and the N-
terminus is a
free amine to aid in coupling, i.e., to cysteine. In one embodiment, a cancer
protein of the
invention is conjugated to an immunogenic agent as discussed herein. In one
embodiment, the cancer protein is conjugated to BSA. The peptides of the
invention, e.g.,
of preferred lengths, can be linked to each other or to other amino acids to
create a longer
peptide/protein. The modulatory peptides can be digests of naturally occurring
proteins
as is outlined above, random peptides, or "biased" random peptides. In a
preferred
embodiment, peptide/protein-based modulators are antibodies, and fragments
thereof, as
defined herein.
[00115] The term "monoclonal antibody", as used herein, refers to an antibody
obtained from a population of substantially homogeneous antibodies, i.e., the
individual
antibodies comprising the population are identical except for possible
naturally occurring
mutations that may be present in minor amounts. Monoclonal antibodies are
highly

34


CA 02754531 2011-09-02
WO 2010/111018 PCT/US2010/026429
specific, being directed against a single antigenic epitope. In contrast,
conventional
(polyclonal) antibody preparations typically include a multitude of antibodies
directed
against (or specific for) different epitopes. In one embodiment, the
polyclonal antibody
contains a plurality of monoclonal antibodies with different epitope
specificities,
affinities, or avidities within a single antigen that contains multiple
antigenic epitopes.
The modifier "monoclonal" indicates the character of the antibody as being
obtained from
a substantially homogeneous population of antibodies, and is not to be
construed as
requiring production of the antibody by any particular method. For example,
the
monoclonal antibodies to be used in accordance with the present invention may
be made
by the hybridoma method first described by Kohler et al., Nature 256: 495
(1975), or may
be made by recombinant DNA methods (see, e.g., U.S. Pat. No. 4,816,567). The
"monoclonal antibodies" may also be isolated from phage antibody libraries
using the
techniques described in Clackson et al., Nature 352: 624-628 (1991) and Marks
et al., J.
Mol. Biol. 222: 581-597 (1991), for example. These monoclonal antibodies will
usually
bind with at least a Kd of about 1 M, more usually at least about 300 nM,
typically at
least about 30 nM, preferably at least about 10 nM, more preferably at least
about 3 nM
or better, usually determined by ELISA.
[00116] A "pharmaceutical excipient" comprises a material such as an adjuvant,
a
carrier, pH-adjusting and buffering agents, tonicity adjusting agents, wetting
agents,
preservative, and the like.
[00117] "Pharmaceutically acceptable" refers to a non-toxic, inert, and/or
composition
that is physiologically compatible with humans or other mammals.
[00118] The term "polynucleotide" means a polymeric form of nucleotides of at
least
bases or base pairs in length, either ribonucleotides or deoxynucleotides or a
modified
form of either type of nucleotide, and is meant to include single and double
stranded
forms of DNA and/or RNA. In the art, this term if often used interchangeably
with
"oligonucleotide". A polynucleotide can comprise a nucleotide sequence
disclosed herein
wherein thymidine (T), as shown for example in Figure 1, can also be uracil
(U); this
definition pertains to the differences between the chemical structures of DNA
and RNA,
in particular the observation that one of the four major bases in RNA is
uracil (U) instead
of thymidine (T).



CA 02754531 2011-09-02
WO 2010/111018 PCT/US2010/026429
[00119] The term "polypeptide" means a polymer of at least about 4, 5, 6, 7,
or 8
amino acids. Throughout the specification, standard three letter or single
letter
designations for amino acids are used. In the art, this term is often used
interchangeably
with "peptide" or "protein".
[00120] A "recombinant" DNA or RNA molecule is a DNA or RNA molecule that has
been subjected to molecular manipulation in vitro.
[00121] As used herein, the term "single-chain Fv" or "scFv" or "single chain"
antibody refers to antibody fragments comprising the VH and VL domains of
antibody,
wherein these domains are present in a single polypeptide chain. Generally,
the Fv
polypeptide further comprises a polypeptide linker between the VH and VL
domains
which enables the sFv to form the desired structure for antigen binding. For a
review of
sFv, see Pluckthun, THE PHARMACOLOGY OF MONOCLONAL ANTIBODIES, vol.
113, Rosenburg and Moore eds. Springer-Verlag, New York, pp. 269-315 (1994).
[00122] As used herein, the terms "specific", "specifically binds" and "binds
specifically" refer to the selective binding of the antibody to the target
antigen epitope.
Antibodies can be tested for specificity of binding by comparing binding to
appropriate
antigen to binding to irrelevant antigen or antigen mixture under a given set
of conditions.
If the antibody binds to the appropriate antigen at least 2, 5, 7, and
preferably 10 times
more than to irrelevant antigen or antigen mixture then it is considered to be
specific. In
one embodiment, a specific antibody is one that only binds the 24P4C12
antigen, but does
not bind to the irrelevent antigen. In another embodiment, a specific antibody
is one that
binds human 24P4C12 antigen but does not bind a non-human 24P4C12 antigen with
70%, 75%, 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% or
greater amino acid homology with the 24P4C 12 antigen. In another embodiment,
a
specific antibody is one that binds human 24P4C12 antigen and binds murine
24P4C12
antigen, but with a higher degree of binding the human antigen. In another
embodiment,
a specific antibody is one that binds human 24P4C12 antigen and binds primate
24P4C12
antigen, but with a higher degree of binding the human antigen. In another
embodiment,
the specific antibody binds to human 24P4C12 antigen and any non-human 24P4C12
antigen, but with a higher degree of binding the human antigen or any
combination
thereof.

36


CA 02754531 2011-09-02
WO 2010/111018 PCT/US2010/026429
[00123] As used herein "to treat" or "therapeutic" and grammatically related
terms,
refer to any improvement of any consequence of disease, such as prolonged
survival, less
morbidity, and/or a lessening of side effects which are the byproducts of an
alternative
therapeutic modality; as is readily appreciated in the art, full eradication
of disease is a
preferred but albeit not a requirement for a treatment act.
[00124] The term "variant" refers to a molecule that exhibits a variation from
a
described type or norm, such as a protein that has one or more different amino
acid
residues in the corresponding position(s) of a specifically described protein
(e.g. the
24P4C12 protein shown in Figure 1.) An analog is an example of a variant
protein.
Splice isoforms and single nucleotides polymorphisms (SNPs) are further
examples of
variants.
[00125] The "24P4C 12-related proteins" of the invention include those
specifically
identified herein (see, Figure 1A - 11), as well as allelic variants,
conservative
substitution variants, analogs and homologs that can be isolated/generated and
characterized without undue experimentation following the methods outlined
herein or
readily available in the art. Fusion proteins that combine parts of different
24P4C12
proteins or fragments thereof, as well as fusion proteins of a 24P4C12 protein
and a
heterologous polypeptide are also included. Such 24P4C12 proteins are
collectively
referred to as the 24P4C 12-related proteins, the proteins of the invention,
or 24P4C 12.
The term "24P4C 12-related protein" refers to a polypeptide fragment or a
24P4C12
protein sequence of 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19,
20, 21, 22, 23,
24, 25, or more than 25 amino acids; or, at least 30, 35, 40, 45, 50, 55, 60,
65, 70, 80, 85,
90, 95, 100, 105, 110, 115, 120, 125, 130, 135, 140, 145, 150, 155, 160, 165,
170, 175,
180, 185, 190, 195, 200, 225, 250, 275, 300, 325, 330, 335, 339 or more amino
acids.
II.) 24P4C12 Antibodies
[00126] Another aspect of the invention provides antibodies that bind to
24P4C12-
related proteins (See Figure 1). Preferred antibodies specifically bind to a
24P4C12-
related protein and do not bind (or bind weakly) to peptides or proteins that
are not
24P4C12-related proteins under physiological conditions. For example,
antibodies that
bind 24P4C12 can bind 24P4C12-related proteins such as 24P4C12 variants and
the
homologs or analogs thereof.

37


CA 02754531 2011-09-02
WO 2010/111018 PCT/US2010/026429
[00127] 24P4C12 antibodies of the invention are particularly useful in cancer
(see,
e.g., Table I) prognostic assays, imaging, and therapeutic methodologies.
Similarly, such
antibodies are useful in the treatment, and/or prognosis of colon and other
cancers, to the
extent 24P4C12 is also expressed or overexpressed in these other cancers.
Moreover,
intracellularly expressed antibodies (e.g., single chain antibodies) are
therapeutically
useful in treating cancers in which the expression of 24P4C12 is involved,
such as
advanced or metastatic colon cancers or other advanced or metastatic cancers.
[00128] Various methods for the preparation of antibodies, specifically
monoclonal
antibodies, are well known in the art. For example, antibodies can be prepared
by
immunizing a suitable mammalian host using a 24P4C12-related protein, peptide,
or
fragment, in isolated or immunoconjugated form (Antibodies: A Laboratory
Manual,
CSH Press, Eds., Harlow, and Lane (1988); Harlow, Antibodies, Cold Spring
Harbor
Press, NY (1989)). In addition, fusion proteins of 24P4C12 can also be used,
such as a
24P4C12 GST-fusion protein. In a particular embodiment, a GST fusion protein
comprising all or most of the amino acid sequence of Figure 1 is produced, and
then used
as an immunogen to generate appropriate antibodies. In another embodiment, a
24P4C12-related protein is synthesized and used as an immunogen.
[00129] In addition, naked DNA immunization techniques known in the art are
used
(with or without purified 24P4C 12-related protein or 24P4C12 expressing
cells) to
generate an immune response to the encoded immunogen (for review, see Donnelly
et al.,
1997, Ann. Rev. Immunol. 15: 617-648).
[00130] The amino acid sequence of a 24P4C12 protein as shown in Figure 1 can
be
analyzed to select specific regions of the 24P4C12 protein for generating
antibodies. For
example, hydrophobicity and hydrophilicity analyses of a 24P4C12 amino acid
sequence
are used to identify hydrophilic regions in the 24P4C12 structure. Regions of
a 24P4C12
protein that show immunogenic structure, as well as other regions and domains,
can
readily be identified using various other methods known in the art, such as
Chou-Fasman,
Garnier-Robson, Kyte-Doolittle, Eisenberg, Karplus-Schultz or Jameson-Wolf
analysis.
Hydrophilicity profiles can be generated using the method of Hopp, T.P. and
Woods,
K.R., 1981, Proc. Natl. Acad. Sci. U.S.A. 78:3824-3828. Hydropathicity
profiles can be
generated using the method of Kyte, J. and Doolittle, R.F., 1982, J. Mol.
Biol. 157:105-
132. Percent (%) Accessible Residues profiles can be generated using the
method of

38


CA 02754531 2011-09-02
WO 2010/111018 PCT/US2010/026429
Janin J., 1979, Nature 277:491-492. Average Flexibility profiles can be
generated using
the method of Bhaskaran R., Ponnuswamy P.K., 1988, Int. J. Pept. Protein Res.
32:242-
255. Beta-turn profiles can be generated using the method of Deleage, G., Roux
B., 1987,
Protein Engineering 1:289-294. Thus, each region identified by any of these
programs or
methods is within the scope of the present invention. Preferred methods for
the
generation of 24P4C12 antibodies are further illustrated by way of the
examples provided
herein. Methods for preparing a protein or polypeptide for use as an immunogen
are well
known in the art. Also well known in the art are methods for preparing
immunogenic
conjugates of a protein with a carrier, such as BSA, KLH or other carrier
protein. In
some circumstances, direct conjugation using, for example, carbodiimide
reagents are
used; in other instances linking reagents such as those supplied by Pierce
Chemical Co.,
Rockford, IL, are effective. Administration of a 24P4C 12 immunogen is often
conducted
by injection over a suitable time period and with use of a suitable adjuvant,
as is
understood in the art. During the immunization schedule, titers of antibodies
can be taken
to determine adequacy of antibody formation.
[00131] 24P4C12 monoclonal antibodies can be produced by various means well
known in the art. For example, immortalized cell lines that secrete a desired
monoclonal
antibody are prepared using the standard hybridoma technology of Kohler and
Milstein or
modifications that immortalize antibody-producing B cells, as is generally
known.
Immortalized cell lines that secrete the desired antibodies are screened by
immunoassay
in which the antigen is a 24P4C12-related protein. When the appropriate
immortalized
cell culture is identified, the cells can be expanded and antibodies produced
either from in
vitro cultures or from ascites fluid.
[00132] The antibodies or fragments of the invention can also be produced by
recombinant means. Regions that bind specifically to the desired regions of a
24P4C12
protein can also be produced in the context of chimeric or complementarity-
determining
region (CDR) grafted antibodies of multiple species origin. Humanized or human
24P4C 12 antibodies can also be produced, and are preferred for use in
therapeutic
contexts. Methods for humanizing murine and other non-human antibodies, by
substituting one or more of the non-human antibody CDRs for corresponding
human
antibody sequences, are well known (see for example, Jones et al., 1986,
Nature 321:
522-525; Riechmann et al., 1988, Nature 332: 323-327; Verhoeyen et al., 1988,
Science

39


CA 02754531 2011-09-02
WO 2010/111018 PCT/US2010/026429
239: 1534-1536). See also, Carter et al., 1993, Proc. Natl. Acad. Sci. USA 89:
4285 and
Sims et al., 1993, J. Immunol. 151: 2296.
[00133] In a preferred embodiment, the antibodies of the present invention
comprise
fully human 24P4C12 antibodies (24P4C12 MAbs). Various methods in the art
provide
means for producing fully human 24P4C12 MAbs. For example, a preferred
embodiment
provides for techniques using transgenic mice, inactivated for antibody
production,
engineered with human heavy and light chains loci referred to as Xenomouse
(Amgen
Fremont, Inc.). An exemplary descritption of preparing transgenic mice that
produce
human antibodies can be found in U.S. 6,657,103. See, also, U.S. Patent Nos.
5,569,825;
5,625,126; 5,633,425; 5,661,016; and 5,545,806; and Mendez, et. al. Nature
Genetics, 15:
146-156 (1998); Kellerman, S.A. & Green, L.L., Curr. Opin. Biotechnol 13, 593-
597
(2002).
[00134] In addition, human antibodies of the invention can be generated using
the
HuMAb mouse (Medarex, Inc.) which contains human immunoglobulin gene miniloci
that encode unrearranged human heavy (mu and gamma) and kappa light chain
immunoglobulin sequences, together with targeted mutations that inactivate the
endogenous mu and kappa chain loci (see e.g., Lonberg, et al. (1994) Nature
368(6474):
856-859).
[00135] In another embodiment, fully human antibodies of the invention can be
raised
using a mouse that carries human immunoglobulin sequences on transgenes and
transchomosomes, such as a mouse that carries a human heavy chain transgene
and a
human light chain transchromosome. Such mice, referred to herein as "KM mice",
such
mice are described in Tomizuka et al. (2000) Proc. Natl. Acad. Sci. USA 97:722-
727 and
PCT Publication WO 02/43478 to Tomizuka, et al.
[00136] Human monoclonal antibodies of the invention can also be prepared
using
phage display methods for screening libraries of human immunoglobulin genes.
Such
phage display methods for isolating human antibodies are established in the
art. See for
example: U.S. Pat. Nos. 5,223,409; 5,403,484; and 5,571,698 to Ladner et al.;
U.S. Pat.
Nos. 5,427,908 and 5,580,717 to Dower et al.; U.S. Pat. Nos. 5,969,108 and
6,172,197 to
McCafferty et al.; and U.S. Pat. Nos. 5,885,793; 6,521,404; 6,544,731;
6,555,313;
6,582,915 and 6,593,081 to Griffiths et al.



CA 02754531 2011-09-02
WO 2010/111018 PCT/US2010/026429
[00137] Human monoclonal antibodies of the invention can also be prepared
using
SCID mice into which human immune cells have been reconstituted such that a
human
antibody response can be generated upon immunization. Such mice are described
in, for
example, U.S. Pat. Nos. 5,476,996 and 5,698,767 to Wilson et al.
[00138] In a preferred embodiment, an 24P4C12 MAbs of the invention comprises
heavy and light chain variable regions of an antibody designated Ha5-1(5)2.1
produced
by a hybridoma deposited under the American Type Culture Collection (ATCC)
Accession No.: PTA-8602 (See, Figure 3), or heavy and light variable regions
comprising amino acid sequences that are homologous to the amino acid
sequences of the
heavy and light chain variable regions of Ha5-1(5)2.1, and wherein the
antibodies retain
the desired functional properties of the 24P4C12 MAbs of the invention. The
heavy
chain variable region of Ha5-1(5)2.1 consists of the amino acid sequence
ranging from
20th Q residue to the 143th S residue of SEQ ID NO: 20, and the light chain
variable
region of Ha5-1(5)2.1 consists of the amino acid sequence ranging from 23th D
residue to
the 130th R residue of SEQ ID NO: 22. As the constant region of the antibody
of the
invention, any subclass of constant region can be chosen. In one embodiment,
human
IgG2 constant region as the heavy chain constant region and human Ig kappa
constant
region as the light chain constant region can be used.
[00139] For example, the invention provides an isolated monoclonal antibody,
or
antigen binding portion thereof, comprising a heavy chain variable region and
a light
chain variable region, wherein:
[00140] (a) the heavy chain variable region comprises an amino acid sequence
that is
at least 80% homologous to heavy chain variable region amino acid sequence set
forth in
Figure 3; and
[00141] (b) the light chain variable region comprises an amino acid sequence
that is at
least 80% homologous to the light chain variable region amino acid sequence
set forth in
Figure 3.
[00142] In other embodiments, the VH and/or VL amino acid sequences may be
85%,
86%,87%,88%,89%,90%,91%,92%,93%,94%, 95%,96%,97%,98% or 99%
homologous to the VH and VL sequences set forth in Figure 3.

41


CA 02754531 2011-09-02
WO 2010/111018 PCT/US2010/026429
[00143] In another embodiment, the invention provides an isolated monoclonal
antibody, or antigen binding portion thereof, comprising a humanized heavy
chain
variable region and a humanized light chain variable region, wherein:
[00144] (a) the heavy chain variable region comprises complementarity
determining
regions (CDRs) having the amino acid sequences of the heavy chain variable
region
CDRs set forth in Figure 3;
[00145] (b) the light chain variable region comprises CDRs having the amino
acid
sequences of the light chain variable region CDRs set forth in Figure 3.
[00146] Engineered antibodies of the invention include those in which
modifications
have been made to framework residues within VH and/or VL (e.g. to improve the
properties of the antibody). Typically such framework modifications are made
to
decrease the immunogenicity of the antibody. For example, one approach is to
"backmutate" one or more framework residues to the corresponding germline
sequence.
More specifically, an antibody that has undergone somatic mutation may contain
framework residues that differ from the germline sequence from which the
antibody is
derived. Such residues can be identified by comparing the antibody framework
sequences to the germline sequences from which the antibody is derived. To
return the
framework region sequences to their germline configuration, the somatic
mutations can
be "backmutated" to the germline sequence by, for example, site-directed
mutagenesis or
PCR-mediated mutagenesis (e.g., "backmutated" from leucine to methionine).
Such
"backmutated" antibodies are also intended to be encompassed by the invention.
[00147] Another type of framework modification involves mutating one or more
residues within the framework region, or even within one or more CDR regions,
to
remove T-cell epitopes to thereby reduce the potential immunogenicity of the
antibody.
This approach is also referred to as "deimmunization" and is described in
further detail in
U.S. Patent Publication No. 2003/0153043 by Can et al.
[00148] In addition or alternative to modifications made within the framework
or CDR
regions, antibodies of the invention may be engineered to include
modifications within
the Fc region, typically to alter one or more functional properties of the
antibody, such as
serum half-life, complement fixation, Fc receptor binding, and/or antigen-
dependent
cellular cytotoxicity. Furthermore, a 24P4C12 MAb of the invention may be
chemically
modified (e.g., one or more chemical moieties can be attached to the antibody)
or be

42


CA 02754531 2011-09-02
WO 2010/111018 PCT/US2010/026429
modified to alter its glycosylation, again to alter one or more functional
properties of the
MAb. Each of these embodiments is described in further detail below.
[00149] In one embodiment, the hinge region of CHI is modified such that the
number
of cysteine residues in the hinge region is altered, e.g., increased or
decreased. This
approach is described further in U.S. Pat. No. 5,677,425 by Bodmer et al. The
number of
cysteine residues in the hinge region of CHI is altered to, for example,
facilitate assembly
of the light and heavy chains or to increase or decrease the stability of the
24P4C12 MAb.
[00150] In another embodiment, the Fc hinge region of an antibody is mutated
to
decrease the biological half life of the 24P4C12 MAb. More specifically, one
or more
amino acid mutations are introduced into the CH2-CH3 domain interface region
of the
Fc-hinge fragment such that the antibody has impaired Staphylococcyl protein A
(SpA)
binding relative to native Fc-hinge domain SpA binding. This approach is
described in
further detail in U.S. Pat. No. 6,165,745 by Ward et al.
[00151] In another embodiment, the 24P4C12 MAb is modified to increase its
biological half life. Various approaches are possible. For example, mutations
can be
introduced as described in U.S. Pat. No. 6,277,375 to Ward. Alternatively, to
increase the
biological half life, the antibody can be altered within the CHI or CL region
to contain a
salvage receptor binding epitope taken from two loops of a CH2 domain of an Fc
region
of an IgG, as described in U.S. Pat. Nos. 5,869,046 and 6,121,022 by Presta et
al.
[00152] In yet other embodiments, the Fc region is altered by replacing at
least one
amino acid residue with a different amino acid residue to alter the effector
function(s) of
the 24P4C12 MAb. For example, one or more amino acids selected from amino acid
specific residues can be replaced with a different amino acid residue such
that the
antibody has an altered affinity for an effector ligand but retains the
antigen-binding
ability of the parent antibody. The effector ligand to which affinity is
altered can be, for
example, an Fc receptor or the Cl component of complement. This approach is
described
in further detail in U.S. Pat. Nos. 5,624,821 and 5,648,260, both by Winter et
al.
[00153] Reactivity of 24P4C12 antibodies with a 24P4C 12-related protein can
be
established by a number of well known means, including Western blot,
immunoprecipitation, ELISA, and FACS analyses using, as appropriate, 24P4C12-
related
proteins, 24P4C12-expressing cells or extracts thereof. A 24P4C12 antibody or
fragment
thereof can be labeled with a detectable marker or conjugated to a second
molecule.

43


CA 02754531 2011-09-02
WO 2010/111018 PCT/US2010/026429
Suitable detectable markers include, but are not limited to, a radioisotope, a
fluorescent
compound, a bioluminescent compound, chemiluminescent compound, a metal
chelator
or an enzyme. Further, bi-specific antibodies specific for two or more 24P4C
12 epitopes
are generated using methods generally known in the art. Homodimeric antibodies
can
also be generated by cross-linking techniques known in the art (e.g., Wolff et
al., Cancer
Res. 53: 2560-2565).
[00154] In yet another preferred embodiment, the 24P4C12 MAb of the invention
is an
antibody comprising heavy and light chain of an antibody designated Ha5-
1(5)2.1. The
heavy chain of Ha5-1(5)2.1 consists of the amino acid sequence ranging from
20th Q
residue to the 469th K residue of SEQ ID NO: 20 and the light chain of Ha5-
1(5)2.1
consists of amino acid sequence ranging from 23t' D residue to the 236th C
residue of
SEQ ID NO: 22 sequence. The sequence of which is set forth in Figure 2 and
Figure 3.
In a preferred embodiment, Ha5-1(5)2.1 is conjugated to a cytotoxic agent.
[00155] The hybridoma producing the antibody designated Ha5-1(5)2.1 was sent
(via
Federal Express) to the American Type Culture Collection (ATCC), P.O. Box
1549,
Manassas, VA 20108 on 08-August-2007 and assigned Accession number PTA-8602.
III.) Antibody-Drug Conjugates Generally
[00156] In another aspect, the invention provides antibody-drug conjugates
(ADCs),
comprising an antibody conjugated to a cytotoxic agent such as a
chemotherapeutic agent,
a drug, a growth inhibitory agent, a toxin (e.g., an enzymatically active
toxin of bacterial,
fungal, plant, or animal origin, or fragments thereof), or a radioactive
isotope (i.e., a
radioconjugate). In another aspect, the invention further provides methods of
using the
ADCs. In one aspect, an ADC comprises any of the above 24P4C12 MAbs covalently
attached to a cytotoxic agent or a detectable agent.
[00157] The use of antibody-drug conjugates for the local delivery of
cytotoxic or
cytostatic agents, i.e. drugs to kill or inhibit tumor cells in the treatment
of cancer
(Syrigos and Epenetos (1999) Anticancer Research 19:605-614; Niculescu-Duvaz
and
Springer (1997) Adv. Drg Del. Rev. 26:151-172; U.S. patent 4,975,278) allows
targeted
delivery of the drug moiety to tumors, and intracellular accumulation therein,
where
systemic administration of these unconjugated drug agents may result in
unacceptable
levels of toxicity to normal cells as well as the tumor cells sought to be
eliminated

44


CA 02754531 2011-09-02
WO 2010/111018 PCT/US2010/026429
(Baldwin et al., (1986) Lancet pp. (Mar. 15, 1986):603-05; Thorpe, (1985)
"Antibody
Carriers Of Cytotoxic Agents In Cancer Therapy: A Review," in Monoclonal
Antibodies
'84: Biological And Clinical Applications, A. Pinchera et al. (ed.s), pp. 475-
506).
Maximal efficacy with minimal toxicity is sought thereby. Both polyclonal
antibodies
and monoclonal antibodies have been reported as useful in these strategies
(Rowland et
al., (1986) Cancer Immunol. Immunother., 21:183-87). Drugs used in these
methods
include daunomycin, doxorubicin, methotrexate, and vindesine (Rowland et al.,
(1986)
supra). Toxins used in antibody-toxin conjugates include bacterial toxins such
as
diphtheria toxin, plant toxins such as ricin, small molecule toxins such as
geldanamycin
(Mandler et al (2000) Jour. of the Nat. Cancer Inst. 92(19):1573-1581; Mandler
et al
(2000) Bioorganic & Med. Chem. Letters 10:1025-1028; Mandler et al (2002)
Bioconjugate Chem. 13:786-791), maytansinoids (EP 1391213; Liu et al., (1996)
Proc.
Natl. Acad. Sci. USA 93:8618-8623), and calicheamicin (Lode et al (1998)
Cancer Res.
58:2928; Hinman et al (1993) Cancer Res. 53:3336-3342). The toxins may effect
their
cytotoxic and cytostatic effects by mechanisms including tubulin binding, DNA
binding,
or topoisomerase inhibition. Some cytotoxic drugs tend to be inactive or less
active when
conjugated to large antibodies or protein receptor ligands.

[00158] Examples of antibody drug conjugates are, ZEVALIN (ibritumomab
tiuxetan, Biogen/Idec) which is an antibody-radioisotope conjugate composed of
a murine
IgGI kappa monoclonal antibody directed against the CD20 antigen found on the
surface
of normal and malignant B lymphocytes and "'In 'In or 90Y radioisotope bound
by a thiourea
linker-chelator (Wiseman et al (2000) Fur. Jour. Nucl. Med. 27(7):766-77;
Wiseman et al
(2002) Blood 99(12):4336-42; Witzig et al (2002) J. Clin. Oncol. 20(10):2453-
63; Witzig
et al (2002) J. Clin. Oncol. 20(15):3262-69).

[00159] Additionally, MYLOTARGTM (gemtuzumab ozogamicin, Wyeth
Pharmaceuticals), an antibody drug conjugate composed of a hu CD33 antibody
linked to
calicheamicin, was approved in 2000 for the treatment of acute myeloid
leukemia by
injection (Drugs of the Future (2000) 25(7):686; US Patent Nos. 4970198;
5079233;
5585089;5606040;5693762;5739116;5767285;5773001).
[00160] In addition, Cantuzumab mertansine (Immunogen, Inc.), an antibody drug
conjugate composed of the huC242 antibody linked via the disulfide linker SPP
to the


CA 02754531 2011-09-02
WO 2010/111018 PCT/US2010/026429
maytansinoid drug moiety, DM1, is advancing into Phase II trials for the
treatment of
cancers that express CanAg, such as colon, pancreatic, gastric, and others.
[00161] Additionally, MLN-2704 (Millennium Pharm., BZL Biologics, Immunogen
Inc.), an antibody drug conjugate composed of the anti-prostate specific
membrane
antigen (PSMA) monoclonal antibody linked to the maytansinoid drug moiety,
DM1, is
under development for the potential treatment of prostate tumors.
[00162] Finally, the auristatin peptides, auristatin E (AE) and
monomethylauristatin
(MMAE), synthetic analogs of dolastatin, were conjugated to chimeric
monoclonal
antibodies cBR96 (specific to Lewis Y on carcinomas) and cAC10 (specific to
CD30 on
hematological malignancies) (Doronina et al (2003) Nature Biotechnology
21(7):778-
784) and are under therapeutic development.
[00163] Further, chemotherapeutic agents useful in the generation of ADCs are
described herein. Enzymatically active toxins and fragments thereof that can
be used
include diphtheria A chain, nonbinding active fragments of diphtheria toxin,
exotoxin A
chain (from Pseudomonas aeruginosa), ricin A chain, abrin A chain, modeccin A
chain,
alpha-sarcin, Aleurites fordii proteins, dianthin proteins, Phytolaca
americana proteins
(PAPI, PAPII, and PAP-S), momordica charantia inhibitor, curcin, crotin,
sapaonaria
officinalis inhibitor, gelonin, mitogellin, restrictocin, phenomycin,
enomycin, and the
tricothecenes. See, e.g., WO 93/21232 published October 28, 1993. A variety of
radionuclides are available for the production of radioconjugated antibodies.
Examples
include 212Bi, 1311, 131In 90Y, and 186Re. Conjugates of the antibody and
cytotoxic agent
are made using a variety of bifunctional protein-coupling agents such as N-
succinimidyl-
3-(2-pyridyldithiol) propionate (SPDP), iminothiolane (IT), bifunctional
derivatives of
imidoesters (such as dimethyl adipimidate HC1), active esters (such as
disuccinimidyl
suberate), aldehydes (such as glutaraldehyde), bis-azido compounds (such as
bis (p-
azidobenzoyl) hexanediamine), bis-diazonium derivatives (such as bis-(p-
diazoniumbenzoyl)-ethylenediamine), diisocyanates (such as toluene 2,6-
diisocyanate),
and bis-active fluorine compounds (such as 1,5-difluoro-2,4-dinitrobenzene).
For
example, a ricin immunotoxin can be prepared as described in Vitetta et al
(1987)
Science, 238:1098. Carbon- 14-labeled 1-isothiocyanatobenzyl-3-
methyldiethylene
triaminepentaacetic acid (MX-DTPA) is an exemplary chelating agent for
conjugation of
radionucleotide to the antibody (W094/11026).

46


CA 02754531 2011-09-02
WO 2010/111018 PCT/US2010/026429
[00164] Conjugates of an antibody and one or more small molecule toxins, such
as a
calicheamicin, maytansinoids, dolastatins, auristatins, a trichothecene, and
CC1065, and
the derivatives of these toxins that have toxin activity, are also
contemplated herein.
III(A). Maytansinoids
[00165] Maytansine compounds suitable for use as maytansinoid drug moieties
are
well known in the art, and can be isolated from natural sources according to
known
methods, produced using genetic engineering techniques (see Yu et al (2002)
PNAS
99:7968-7973), or maytansinol and maytansinol analogues prepared synthetically
according to known methods.
[00166] Exemplary maytansinoid drug moieties include those having a modified
aromatic ring, such as: C-19-dechloro (US 4256746) (prepared by lithium
aluminum
hydride reduction of ansamytocin P2); C-20-hydroxy (or C-20-demethyl) +/-C-19-
dechloro (US Pat. Nos. 4361650 and 4307016) (prepared by demethylation using
Streptomyces or Actinomyces or dechlorination using LAH); and C-20-demethoxy,
C-20-
acyloxy (-OCOR), +/-dechloro (U.S. Pat. No. 4,294,757) (prepared by acylation
using
acyl chlorides). and those having modifications at other positions
[00167] Exemplary maytansinoid drug moieties also include those having
modifications such as: C-9-SH (US 4424219) (prepared by the reaction of
maytansinol
with H2S or P2S5); C-14-alkoxymethyl(demethoxy/CH2 OR)(US 4331598); C-14-
hydroxymethyl or acyloxymethyl (CH2OH or CH2OAc) (US 4450254) (prepared from
Nocardia); C-15-hydroxy/acyloxy (US 4364866) (prepared by the conversion of
maytansinol by Streptomyces); C-15-methoxy (US Pat. Nos. 4313946 and 4315929)
(isolated from Trewia nudlflora); C-18-N-demethyl (US Pat. Nos. 4362663 and
4322348)
(prepared by the demethylation of maytansinol by Streptomyces); and 4,5-deoxy
(US
4371533) (prepared by the titanium trichloride/LAH reduction of maytansinol).
[00168] ADCs containing maytansinoids, methods of making same, and their
therapeutic use are disclosed, for example, in U.S. Patent Nos. 5,208,020;
5,416,064;
6,441163 and European Patent EP 0 425 235 B1, the disclosures of which are
hereby
expressly incorporated by reference. Liu et al., Proc. Natl. Acad. Sci. USA
93:8618-8623
(1996) described ADCs comprising a maytansinoid designated DM1 linked to the
monoclonal antibody C242 directed against human colorectal cancer. The
conjugate was
found to be highly cytotoxic towards cultured colon cancer cells, and showed
antitumor

47


CA 02754531 2011-09-02
WO 2010/111018 PCT/US2010/026429
activity in an in vivo tumor growth assay. Chari et al., Cancer Research
52:127-131
(1992) describe ADCs in which a maytansinoid was conjugated via a disulfide
linker to
the murine antibody A7 binding to an antigen on human colon cancer cell lines,
or to
another murine monoclonal antibody TA.1 that binds the HER-2/neu oncogene. The
cytotoxicity of the TA.1-maytansonoid conjugate was tested in vitro on the
human breast
cancer cell line SK-BR-3, which expresses 3 x 105 HER-2 surface antigens per
cell. The
drug conjugate achieved a degree of cytotoxicity similar to the free
maytansinoid drug,
which could be increased by increasing the number of maytansinoid molecules
per
antibody molecule. The A7-maytansinoid conjugate showed low systemic
cytotoxicity in
mice.
III(B). Auristatins and dolastatins
[00169] In some embodiments, the ADC comprises an antibody of the invention
conjugated to dolastatins or dolostatin peptidic analogs and derivatives, the
auristatins
(US Patent Nos. 5635483; 5780588). Dolastatins and auristatins have been shown
to
interfere with microtubule dynamics, GTP hydrolysis, and nuclear and cellular
division
(Woyke et al (2001) Antimicrob. Agents and Chemother. 45(12):3580-3584) and
have
anticancer (US 5663149) and antifungal activity (Pettit et al (1998)
Antimicrob. Agents
Chemother. 42:2961-2965). The dolastatin or auristatin drug moiety may be
attached to
the antibody through the N (amino) terminus or the C (carboxyl) terminus of
the peptidic
drug moiety (WO 02/088172).
[00170] Exemplary auristatin embodiments include the N-terminus linked
monomethylauristatin drug moieties DE and DF, disclosed in "Senter et al,
Proceedings
of the American Association for Cancer Research, Volume 45, Abstract Number
623,
presented March 28, 2004 and described in United States Patent Publication No.
2005/0238648, the disclosure of which is expressly incorporated by reference
in its
entirety.
[00171] An exemplary auristatin embodiment is MMAE (wherein the wavy line
indicates the covalent attachment to a linker (L) of an antibody drug
conjugate).

O H OH
N,,
,,,. YY
N N N a
N Y r ---
O O\ O O~ O
MMAE
48


CA 02754531 2011-09-02
WO 2010/111018 PCT/US2010/026429
[00172] Another exemplary auristatin embodiment is MMAF, wherein the wavy line
indicates the covalent attachment to a linker (L) of an antibody drug
conjugate (US
2005/0238649):

H O
'-Yl_r \
4 Y
N N
N N
O OINI O O~ O O OH
MMAF
[00173] Additional exemplary embodiments comprising MMAE or MMAF and
various linker components (described further herein) have the following
structures and
abbreviations (wherein Ab means antibody and p is 1 to about 8):

Ab S O H O
O p OWN N N 0,X H
N~VaI Cit-N" O O O 0,0 i
H O ~'
O P
Ab-MC-vc-PAB-MMAF

Ab-S O H O OH
O O OWN N N H
( 1 0 0', 0
N`~~Val-Cit-N" v O O
O H P
Ab-MC-vc-PAB-MMAE
Ab-S
O
O H H
N N N N N
O I O I O,~ O 0,0
O OH i
Ab-MC-MMAF
[00174] Typically, peptide-based drug moieties can be prepared by forming a
peptide
bond between two or more amino acids and/or peptide fragments. Such peptide
bonds
can be prepared, for example, according to the liquid phase synthesis method
(see E.
Schroder and K. Liibke, "The Peptides", volume 1, pp 76-136, 1965, Academic
Press)
that is well known in the field of peptide chemistry. The
auristatin/dolastatin drug
moieties may be prepared according to the methods of: US 5635483; US 5780588;
Pettit
et al (1989) J. Am. Chem. Soc. 111:5463-5465; Pettit et al (1998) Anti-Cancer
Drug

49


CA 02754531 2011-09-02
WO 2010/111018 PCT/US2010/026429
Design 13:243-277; Pettit, G.R., et al. Synthesis, 1996, 719-725; Pettit et al
(1996) J.
Chem. Soc. Perkin Trans. 1 5:859-863; and Doronina (2003) Nat Biotechnol
21(7):778-
784.

111(C). Calicheamicin
[00175] In other embodiments, the ADC comprises an antibody of the invention
conjugated to one or more calicheamicin molecules. The calicheamicin family of
antibiotics are capable of producing double-stranded DNA breaks at sub-
picomolar
concentrations. For the preparation of conjugates of the calicheamicin family,
see U.S.
patents 5,712,374, 5,714,586, 5,739,116, 5,767,285, 5,770,701, 5,770,710,
5,773,001,
5,877,296 (all to American Cyanamid Company). Structural analogues of
calicheamicin
which may be used include, but are not limited to, yii, a2', a3 N-acetyl-yii,
PSAG and O'
(Hinman et al., Cancer Research 53:3336-3342 (1993), Lode et al., Cancer
Research
58:2925-2928 (1998) and the aforementioned U.S. patents to American Cyanamid).
Another anti-tumor drug that the antibody can be conjugated is QFA which is an
antifolate. Both calicheamicin and QFA have intracellular sites of action and
do not
readily cross the plasma membrane. Therefore, cellular uptake of these agents
through
antibody mediated internalization greatly enhances their cytotoxic effects.

III(D). Other Cytotoxic Agents
[00176] Other antitumor agents that can be conjugated to the antibodies of the
invention include BCNU, streptozoicin, vincristine and 5-fluorouracil, the
family of
agents known collectively LL-E33288 complex described in U.S. patents
5,053,394,
5,770,710, as well as esperamicins (U.S. patent 5,877,296).
[00177] Enzymatically active toxins and fragments thereof which can be used
include
diphtheria A chain, nonbinding active fragments of diphtheria toxin, exotoxin
A chain
(from Pseudomonas aeruginosa), ricin A chain, abrin A chain, modeccin A chain,
alpha-
sarcin, Aleurites fordii proteins, dianthin proteins, Phytolaca americana
proteins (PAPI,
PAPII, and PAP-S), momordica charantia inhibitor, curcin, crotin, sapaonaria
officinalis
inhibitor, gelonin, mitogellin, restrictocin, phenomycin, enomycin and the
tricothecenes.
See, for example, WO 93/21232 published October 28, 1993.
[00178] The present invention further contemplates an ADC formed between an
antibody and a compound with nucleolytic activity (e.g., a ribonuclease or a
DNA
endonuclease such as a deoxyribonuclease; DNase).



CA 02754531 2011-09-02
WO 2010/111018 PCT/US2010/026429
[00179] For selective destruction of the tumor, the antibody may comprise a
highly
radioactive atom. A variety of radioactive isotopes are available for the
production of
radioconjugated antibodies. Examples include At211 I131 1125 Y90 Rei86, Re'88,
Sm'53
Bi212 P32, Pb212 and radioactive isotopes of Lu. When the conjugate is used
for detection,
it may comprise a radioactive atom for scintigraphic studies, for example
tc99m or I123, or a
spin label for nuclear magnetic resonance (NMR) imaging (also known as
magnetic
resonance imaging, mri), such as iodine- 123 again, iodine-131, indium-111,
fluorine- 19,
carbon-13, nitrogen-15, oxygen-17, gadolinium, manganese or iron.
[00180] The radio- or other labels may be incorporated in the conjugate in
known
ways. For example, the peptide may be biosynthesized or may be synthesized by
chemical amino acid synthesis using suitable amino acid precursors involving,
for
example, fluorine-19 in place of hydrogen. Labels such as tc99iT' or I123'.
Re'86, Re188 and

In"' can be attached via a cysteine residue in the peptide. Yttrium-90 can be
attached via
a lysine residue. The IODOGEN method (Fraker et al (1978) Biochem. Biophys.
Res.
Commun. 80: 49-57 can be used to incorporate iodine-123. "Monoclonal
Antibodies in
Immunoscintigraphy" (Chatal,CRC Press 1989) describes other methods in detail.

IV.) Antibody-Drug Conjugate Compounds which bind 24P4C12
[00181] The present invention provides, inter alia, antibody-drug conjugate
compounds for targeted delivery of drugs. The inventors have made the
discovery that
the antibody-drug conjugate compounds have potent cytotoxic and/or cytostatic
activity
against cells expressing 24P4C12. The antibody-drug conjugate compounds
comprise an
Antibody unit covalently linked to at least one Drug unit. The Drug units can
be
covalently linked directly or via a Linker unit (-LU-).
[00182] In some embodiments, the antibody drug conjugate compound has the
following formula:
L - (LU-D)p (I)
or a pharmaceutically acceptable salt or solvate thereof; wherein:
L is the Antibody unit, e.g., 24P4C12 MAb of the present invention, and
(LU-D) is a Linker unit-Drug unit moiety, wherein:
LU- is a Linker unit, and

51


CA 02754531 2011-09-02
WO 2010/111018 PCT/US2010/026429
-D is a drug unit having cytostatic or cytotoxic activity against a target
cell; and
p is an integer from 1 to 20.
[00183] In some embodiments, p ranges from 1 to 10, 1 to 9, 1 to 8, 1 to 7, 1
to 6, 1 to
5, 1 to 4, 1 to 3, or 1 to 2. In some embodiments, p ranges from 2 to 10, 2 to
9, 2 to 8, 2
to 7, 2 to 6, 2 to 5, 2 to 4 or 2 to 3. In other embodiments, p is 1, 2, 3, 4,
5 or 6. In some
embodiments, p is 2 or 4.
[00184] In some embodiments, the antibody drug conjugate compound has the
following formula:
L - (Aa Ww-Yy D)p (II)
or a pharmaceutically acceptable salt or solvate thereof, wherein:
L is the Antibody unit, e.g., 24P4C12 MAb; and
-Aa Ww Yy- is a Linker unit (LU), wherein:
-A- is a Stretcher unit,
a is 0 or 1,
each -W- is independently an Amino Acid unit,
w is an integer ranging from 0 to 12,
-Y- is a self-immolative spacer unit,
y is 0, 1 or 2;
-D is a drug units having cytostatic or cytotoxic activity against the target
cell; and
p is an integer from 1 to 20.
[00185] In some embodiments, a is 0 or 1, w is 0 or 1, and y is 0, 1 or 2. In
some
embodiments, a is 0 or 1, w is 0 or 1, and y is 0 or 1. In some embodiments, p
ranges
from Ito10,1to9,1to8,1to7,1to6,1to5,1to4,1to3,orlto2.Insome
embodiments, p ranges from 2 to 8, 2 to 7, 2 to 6, 2 to 5, 2 to 4 or 2 to 3.
In other
embodiments, p is 1, 2, 3, 4, 5 or 6. In some embodiments, p is 2 or 4. In
some
embodiments, when w is not zero, y is 1 or 2. In some embodiments, when w is 1
to 12, y
is 1 or 2. In some embodiments, w is 2 to 12 and y is 1 or 2. In some
embodiments, a is
1 and w and y are 0.
[00186] For compositions comprising a plurality antibodies, the drug loading
is
represented by p, the average number of drug molecules per Antibody. Drug
loading may
52


CA 02754531 2011-09-02
WO 2010/111018 PCT/US2010/026429
range from 1 to 20 drugs (D) per Antibody. The average number of drugs per
antibody in
preparation of conjugation reactions may be characterized by conventional
means such as
mass spectroscopy, ELISA assay, and HPLC. The quantitative distribution of
Antibody-
Drug-Conjugates in terms of p may also be determined. In some instances,
separation,
purification, and characterization of homogeneous Antibody-Drug-conjugates
where p is
a certain value from Antibody-Drug-Conjugates with other drug loadings may be
achieved by means such as reverse phase HPLC or electrophoresis. In exemplary
embodiments, p is from 2 to 8.
[00187] The generation of Antibody-drug conjugate compounds can be
accomplished
by any technique known to the skilled artisan. Briefly, the Antibody-drug
conjugate
compounds comprise 24P4C 12 MAb as the Antibody unit, a drug, and optionally a
linker
that joins the drug and the binding agent. In a preferred embodiment, the
Antibody is
24P4C 12 MAb comprising heavy and light chain variable regions of an antibody
designated Ha5-1(5)2.1 described above. In more preferred embodiment, the
Antibody is
24P4C12 MAb comprising heavy and light chain of an antibody designated Ha5-
1(5)2.1
described above. A number of different reactions are available for covalent
attachment of
drugs and/or linkers to binding agents. This is often accomplished by reaction
of the
amino acid residues of the binding agent, e.g., antibody molecule, including
the amine
groups of lysine, the free carboxylic acid groups of glutamic and aspartic
acid, the
sulfhydryl groups of cysteine and the various moieties of the aromatic amino
acids. One
of the most commonly used non-specific methods of covalent attachment is the
carbodiimide reaction to link a carboxy (or amino) group of a compound to
amino (or
carboxy) groups of the antibody. Additionally, bifunctional agents such as
dialdehydes or
imidoesters have been used to link the amino group of a compound to amino
groups of an
antibody molecule. Also available for attachment of drugs to binding agents is
the Schiff
base reaction. This method involves the periodate oxidation of a drug that
contains glycol
or hydroxy groups, thus forming an aldehyde which is then reacted with the
binding
agent. Attachment occurs via formation of a Schiff base with amino groups of
the
binding agent. Isothiocyanates can also be used as coupling agents for
covalently
attaching drugs to binding agents. Other techniques are known to the skilled
artisan and
within the scope of the present invention.

53


CA 02754531 2011-09-02
WO 2010/111018 PCT/US2010/026429
[00188] In certain embodiments, an intermediate, which is the precursor of the
linker,
is reacted with the drug under appropriate conditions. In certain embodiments,
reactive
groups are used on the drug and/or the intermediate. The product of the
reaction between
the drug and the intermediate, or the derivatized drug, is subsequently
reacted with the
24P4C12 MAb under appropriate conditions.
[00189] Each of the particular units of the Antibody-drug conjugate compounds
is
described in more detail herein. The synthesis and structure of exemplary
Linker units,
Stretcher units, Amino Acid units, self-immolative Spacer unit, and Drug units
are also
described in U.S. Patent Application Publication Nos. 2003-0083263, 2005-
0238649 and
2005-0009751, each if which is incorporated herein by reference in its
entirety and for all
purposes.

V.) Linker Units
[00190] Typically, the antibody-drug conjugate compounds comprise a Linker
unit
between the drug unit and the antibody unit. In some embodiments, the linker
is
cleavable under intracellular conditions, such that cleavage of the linker
releases the drug
unit from the antibody in the intracellular environment. In yet other
embodiments, the
linker unit is not cleavable and the drug is released, for example, by
antibody degradation.
[00191] In some embodiments, the linker is cleavable by a cleaving agent that
is
present in the intracellular environment (e.g., within a lysosome or endosome
or
caveolea). The linker can be, e.g., a peptidyl linker that is cleaved by an
intracellular
peptidase or protease enzyme, including, but not limited to, a lysosomal or
endosomal
protease. In some embodiments, the peptidyl linker is at least two amino acids
long or at
least three amino acids long. Cleaving agents can include cathepsins B and D
and
plasmin, all of which are known to hydrolyze dipeptide drug derivatives
resulting in the
release of active drug inside target cells (see, e.g., Dubowchik and Walker,
1999, Pharm.
Therapeutics 83:67-123). Most typical are peptidyl linkers that are cleavable
by enzymes
that are present in 24P4C12-expressing cells. For example, a peptidyl linker
that is
cleavable by the thiol-dependent protease cathepsin-B, which is highly
expressed in
cancerous tissue, can be used (e.g., a Phe-Leu or a Gly-Phe-Leu-Gly linker
(SEQ ID NO:
25)). Other examples of such linkers are described, e.g., in U.S. Patent No.
6,214,345,
incorporated herein by reference in its entirety and for all purposes. In a
specific

54


CA 02754531 2011-09-02
WO 2010/111018 PCT/US2010/026429
embodiment, the peptidyl linker cleavable by an intracellular protease is a
Val-Cit linker
or a Phe-Lys linker (see, e.g., U.S. Patent 6,214,345, which describes the
synthesis of
doxorubicin with the val-cit linker). One advantage of using intracellular
proteolytic
release of the therapeutic agent is that the agent is typically attenuated
when conjugated
and the serum stabilities of the conjugates are typically high.
[00192] In other embodiments, the cleavable linker is pH-sensitive, i.e.,
sensitive to
hydrolysis at certain pH values. Typically, the pH-sensitive linker
hydrolyzable under
acidic conditions. For example, an acid-labile linker that is hydrolyzable in
the lysosome
(e.g., a hydrazone, semicarbazone, thiosemicarbazone, cis-aconitic amide,
orthoester,
acetal, ketal, or the like) can be used. (See, e.g., U.S. Patent Nos.
5,122,368; 5,824,805;
5,622,929; Dubowchik and Walker, 1999, Pharm. Therapeutics 83:67-123; Neville
et al.,
1989, Biol. Chem. 264:14653-14661.) Such linkers are relatively stable under
neutral pH
conditions, such as those in the blood, but are unstable at below pH 5.5 or
5.0, the
approximate pH of the lysosome. In certain embodiments, the hydrolyzable
linker is a
thioether linker (such as, e.g., a thioether attached to the therapeutic agent
via an
acylhydrazone bond (see, e.g., U.S. Patent No. 5,622,929).
[00193] In yet other embodiments, the linker is cleavable under reducing
conditions
(e.g., a disulfide linker). A variety of disulfide linkers are known in the
art, including, for
example, those that can be formed using SATA (N-succinimidyl-S-
acetylthioacetate),
SPDP (N-succinimidyl-3-(2-pyridyldithio)propionate), SPDB (N-succinimidyl-3-(2-

pyridyldithio)butyrate) and SMPT (N-succinimidyl-oxycarbonyl-alpha-methyl-
alpha-(2-
pyridyl-dithio)toluene), SPDB and SMPT. (See, e.g., Thorpe et al., 1987,
Cancer Res.
47:5924-593 1; Wawrzynczak et al., In Immunoconjugates: Antibody Conjugates in
Radioimagery and Therapy of Cancer (C. W. Vogel ed., Oxford U. Press, 1987.
See also
U.S. Patent No. 4,880,935.)
[00194] In yet other specific embodiments, the linker is a malonate linker
(Johnson et
al., 1995, Anticancer Res. 15:1387-93), a maleimidobenzoyl linker (Lau et al.,
1995,
Bioorg-Med-Chem. 3(10):1299-1304), or a 3'-N-amide analog (Lau et al., 1995,
Bioorg-
Med-Chem. 3(10):1305-12).
[00195] In yet other embodiments, the linker unit is not cleavable and the
drug is
released by antibody degradation. (See U.S. Publication No. 2005/0238649
incorporated
by reference herein in its entirety and for all purposes).



CA 02754531 2011-09-02
WO 2010/111018 PCT/US2010/026429
[00196] Typically, the linker is not substantially sensitive to the
extracellular
environment. As used herein, "not substantially sensitive to the extracellular
environment," in the context of a linker, means that no more than about 20%,
typically no
more than about 15%, more typically no more than about 10%, and even more
typically
no more than about 5%, no more than about 3%, or no more than about 1% of the
linkers,
in a sample of antibody-drug conjugate compound, are cleaved when the antibody-
drug
conjugate compound presents in an extracellular environment (e.g., in plasma).
Whether
a linker is not substantially sensitive to the extracellular environment can
be determined,
for example, by incubating with plasma the antibody-drug conjugate compound
for a
predetermined time period (e.g., 2, 4, 8, 16, or 24 hours) and then
quantitating the amount
of free drug present in the plasma.
[00197] In other, non-mutually exclusive embodiments, the linker promotes
cellular
internalization. In certain embodiments, the linker promotes cellular
internalization when
conjugated to the therapeutic agent (i.e., in the milieu of the linker-
therapeutic agent
moiety of the antibody-drug conjugate compound as described herein). In yet
other
embodiments, the linker promotes cellular internalization when conjugated to
both the
auristatin compound and the 24P4C12 MAb.
[00198] A variety of exemplary linkers that can be used with the present
compositions
and methods are described in WO 2004-010957, U.S. Publication No.
2006/0074008,
U.S. Publication No. 20050238649, and U.S. Publication No. 2006/0024317 (each
of
which is incorporated by reference herein in its entirety and for all
purposes).
[00199] A "Linker unit" (LU) is a bifunctional compound that can be used to
link a
Drug unit and a Antibody unit to form an antibody-drug conjugate compound. In
some
embodiments, the Linker unit has the formula:
-Aa-Ww Yy-
wherein:-A- is a Stretcher unit,
a is 0 or 1,
each -W- is independently an Amino Acid unit,
w is an integer ranging from 0 to 12,
-Y- is a self-immolative Spacer unit, and
yis0,1or2.

56


CA 02754531 2011-09-02
WO 2010/111018 PCT/US2010/026429
[00200] In some embodiments, a is 0 or 1, w is 0 or 1, and y is 0, 1 or 2. In
some
embodiments, a is 0 or 1, w is 0 or 1, and y is 0 or 1. In some embodiments,
when w is 1
to 12, y is 1 or 2. In some embodiments, w is 2 to 12 and y is 1 or 2. In some
embodiments, a is 1 and w and y are 0.

VI.) The Stretcher Unit
[00201] The Stretcher unit ( A ), when present, is capable of linking an
Antibody unit
to an Amino Acid unit (-W-), if present, to a Spacer unit (-Y-), if present;
or to a Drug
unit (-D). Useful functional groups that can be present on a 24P4C12 MAb (e.g.
Ha5-
1(5)2.1), either naturally or via chemical manipulation include, but are not
limited to,
sulfhydryl, amino, hydroxyl, the anomeric hydroxyl group of a carbohydrate,
and
carboxyl. Suitable functional groups are sulfhydryl and amino. In one example,
sulfhydryl groups can be generated by reduction of the intramolecular
disulfide bonds of
a 24P4C12 MAb. In another embodiment, sulfhydryl groups can be generated by
reaction of an amino group of a lysine moiety of a 24P4C12 MAb with 2-
iminothiolane
(Traut's reagent) or other sulfhydryl generating reagents. In certain
embodiments, the
24P4C12 MAb is a recombinant antibody and is engineered to carry one or more
lysines.
In certain other embodiments, the recombinant 24P4C12 MAb is engineered to
carry
additional sulfhydryl groups, e.g., additional cysteines.
[00202] In one embodiment, the Stretcher unit forms a bond with a sulfur atom
of the
Antibody unit. The sulfur atom can be derived from a sulfhydryl group of an
antibody.
Representative Stretcher units of this embodiment are depicted within the
square brackets
of Formulas IIIa and I1Ib, wherein L-, -W-, -Y-, -D, w and y are as defined
above, and R'7
is selected from -Ci-Cio alkylene-, -Ci-Cio alkenylene-, -Ci-Cio alkynylene-,
carbocyclo-,
-O-(CI-C8 alkylene)-, O-(CI-C8 alkenylene)-, -O-(CI-C8 alkynylene)-, -arylene-
, -Ci-Cio
alkylene-arylene-, -C2-Cio alkenylene-arylene, -C2-Cio alkynylene-arylene, -
arylene-Ci-
Cio alkylene-, -arylene-C2-Cio alkenylene-, -arylene-C2-Cio alkynylene-, -Ci-
Cio
alkylene-(carbocyclo)-, -C2-Cio alkenylene-( carbocyclo)-,
-C2-Cio alkynylene-(carbocyclo)-, -(carbocyclo)-Ci-Cio alkylene-, -
(carbocyclo)-C2-Cio
alkenylene-, -(carbocyclo)-C2-Cio alkynylene, -heterocyclo-, -Ci-Cio alkylene-
(heterocyclo)-,
-C2-Cio alkenylene-(heterocyclo)-, -C2-CIO alkynylene-(heterocyclo)-, -
(heterocyclo)-Ci-
57


CA 02754531 2011-09-02
WO 2010/111018 PCT/US2010/026429
C10 alkylene-, -( heterocyclo)-C2-Cio alkenylene-, -( heterocyclo)-Ci-Cio
alkynylene-, -
(CH2CH2O)r , or -(CH2CH2O)r CH2-, and r is an integer ranging from 1-10,
wherein said
alkyl, alkenyl, alkynyl, alkylene, alkenylene, alkynyklene, aryl, carbocycle,
carbocyclo,
heterocyclo, and arylene radicals, whether alone or as part of another group,
are
optionally substituted. In some embodiments, said alkyl, alkenyl, alkynyl,
alkylene,
alkenylene, alkynyklene, aryl, carbocyle, carbocyclo, heterocyclo, and arylene
radicals,
whether alone or as part of another group, are unsubstituted. In some
embodiments, R'7
is selected from -CI-C10 alkylene-, - carbocyclo-, -O-(C1-C8 alkylene)-,
-arylene-, -CI-C10 alkylene-arylene-, -arylene-Ci-Cio alkylene-, -CI-C10
alkylene-
(carbocyclo)-,
-( carbocyclo)-Ci-C10 alkylene-, -C3-C8 heterocyclo-, -CI-C10 alkylene-(
heterocyclo)-, -(
heterocyclo)-Ci-Cio alkylene-, -(CH2CH2O)r, and -(CH2CH2O)rCH2-; and r is an
integer
ranging from 1-10, wherein said alkylene groups are unsubstituted and the
remainder of
the groups are optionally substituted.
[00203] It is to be understood from all the exemplary embodiments that even
where not
denoted expressly, from 1 to 20 drug moieties can be linked to an Antibody (p
= 1-20).

O
N-R17-C(O) WW Yy--D
O
[00204] IIIa
L H2 CONH-R17-C(O) Ww Yy-D
[00205] IIIb
[00206] An illustrative Stretcher unit is that of Formula IIIa wherein R'7 is -
(CH2)5-:
O

N
O
[00207] 0
[00208] Another illustrative Stretcher unit is that of Formula IIIa wherein
R'7
is -(CH2CH2O)r CH2-; and r is 2:

58


CA 02754531 2011-09-02
WO 2010/111018 PCT/US2010/026429
O

N---~ON /\O

O
[00209] 0
[00210] An illustrative Stretcher unit is that of Formula Isla wherein R'7 is
-arylene- or arylene-Ci-Cio alkylene-. In some embodiments, the aryl group is
an
unsubstituted phenyl group.
[00211] Still another illustrative Stretcher unit is that of Formula IIIb
wherein R'7 is -
(CH2)s-:
O
AIA N
[00212] 0
[00213] In certain embodiments, the Stretcher unit is linked to the Antibody
unit via a
disulfide bond between a sulfur atom of the Antibody unit and a sulfur atom of
the
Stretcher unit. A representative Stretcher unit of this embodiment is depicted
within the
square brackets of Formula IV, wherein R'7, L-, -W-, -Y-, -D, w and y are as
defined
above.

L=S S-R17-C(O) Ww Yy-D

[00214] IV
[00215] It should be noted that throughout this application, the S moiety in
the formula
below refers to a sulfur atom of the Antibody unit, unless otherwise indicated
by context.
L S- -
[00216]
[00217] In yet other embodiments, the Stretcher contains a reactive site that
can form a
bond with a primary or secondary amino group of an Antibody. Examples of these
reactive sites include, but are not limited to, activated esters such as
succinimide esters, 4
nitrophenyl esters, pentafluorophenyl esters, tetrafluorophenyl esters,
anhydrides, acid
chlorides, sulfonyl chlorides, isocyanates and isothiocyanates. Representative
Stretcher
units of this embodiment are depicted within the square brackets of Formulas
Va and Vb,
wherein -R'7-, L-, -W-, -Y-, -D, w and y are as defined above;

59


CA 02754531 2011-09-02
WO 2010/111018 PCT/US2010/026429

L C(O)NH-R17-C(O) WW-Yy- D
[00218] Va
S
C
L .NH-R17-C(O) Ww Yy-D

[00219] Vb
[00220] In some embodiments, the Stretcher contains a reactive site that is
reactive to a
modified carbohydrate's (-CHO) group that can be present on an Antibody. For
example,
a carbohydrate can be mildly oxidized using a reagent such as sodium periodate
and the
resulting (-CHO) unit of the oxidized carbohydrate can be condensed with a
Stretcher that
contains a functionality such as a hydrazide, an oxime, a primary or secondary
amine, a
hydrazine, a thiosemicarbazone, a hydrazine carboxylate, and an arylhydrazide
such as
those described by Kaneko et al., 1991, Bioconjugate Chem. 2:133-41.
Representative
Stretcher units of this embodiment are depicted within the square brackets of
Formulas
VIa, VIb, and VIc, wherein -R'7-, L-, -W-, -Y-, -D, w and y are as defined as
above.

L N-NH-R17-C(0) WW-Yy-D

[00221] VIa
L N-0-R17-C(0) WW-Yy-D

[00222] VIb
0

L N-NH-IC R17-C(O) Ww-Yy-D

[00223] VIC


CA 02754531 2011-09-02
WO 2010/111018 PCT/US2010/026429
VII.) The Amino Acid Unit
[00224] The Amino Acid unit (-W-), when present, links the Stretcher unit to
the
Spacer unit if the Spacer unit is present, links the Stretcher unit to the
Drug moiety if the
Spacer unit is absent, and links the Antibody unit to the Drug unit if the
Stretcher unit and
Spacer unit are absent.
[00225] Ww can be, for example, a monopeptide, dipeptide, tripeptide,
tetrapeptide,
pentapeptide, hexapeptide, heptapeptide, octapeptide, nonapeptide,
decapeptide,
undecapeptide or dodecapeptide unit. Each -W- unit independently has the
formula
denoted below in the square brackets, and w is an integer ranging from 0 to
12:

[cH3

N O 0 -- i

R19
[00226] Rig , or

[00227] wherein R19 is hydrogen, methyl, isopropyl, isobutyl, sec-butyl,
benzyl, p-
hydroxybenzyl, -CH2OH, -CH(OH)CH3, -CH2CH2SCH3, -CH2CONH2, -CH2COOH,
-CH2CH2CONH2, -CH2CH2COOH, -(CH2)3NHC(=NH)NH2, -(CH2)3NH2,
-(CH2)3NHCOCH3, -(CH2)3NHCHO, -(CH2)4NHC(=NH)NH2, -(CH2)4NH2,
-(CH2)4NHCOCH3, -(CH2)4NHCHO, -(CH2)3NHCONH2, -(CH2)4NHCONH2,
-CH2CH2CH(OH)CH2NH2, 2-pyridylmethyl-, 3-pyridylmethyl-, 4-pyridylmethyl-,
phenyl, cyclohexyl,

61


CA 02754531 2011-09-02
WO 2010/111018 PCT/US2010/026429
OH
00

00
\ \ I $ CH2 OJ or CH

N
[00228] H
[00229] In some embodiments, the Amino Acid unit can be enzymatically cleaved
by
one or more enzymes, including a cancer or tumor-associated protease, to
liberate the
Drug unit (-D), which in one embodiment is protonated in vivo upon release to
provide a
Drug (D).
[00230] In certain embodiments, the Amino Acid unit can comprise natural amino
acids. In other embodiments, the Amino Acid unit can comprise non-natural
amino acids.
Illustrative Ww units are represented by formulas (VII)-(IX):
O R21
H
\/N
N
H
[00231] R20 0 (VII)
[00232] wherein R20 and R21 are as follows:
R20 R21

Benzyl (CH2)4NH2;
methyl (CH2)4NH2;
isopropyl (CH2)4NH2;
isopropyl (CH2)3NHCONH2;

62


CA 02754531 2011-09-02
WO 2010/111018 PCT/US2010/026429
benzyl (CH2)3NHCONH2;
isobutyl (CH2)3NHCONH2;
sec-butyl (CH2)3NHCONH2;

CH (CH2)3NHCONH2;
CNO
H
benzyl methyl;
benzyl (CH2)3NHC(=NH)NH2;
[00233]
O R21 O
1-1 __,y N N S
N
H
[00234] R20 O R22 (VIII)
[00235] wherein R20, R21 and R22 are as follows:
R20 R21 R22

benzyl benzyl (CH2)4NH2;
isopropyl benzyl (CH2)4NH2; and
H benzyl (CH2)4NH2;

O R21 O R23
__,y N
/N N
`L
`? H H
[00236] R20 o R22 0 (IX)
[00237] wherein R20, R21, R22 and R23 are as follows:
R20 R21 R22 R23

H benzyl isobutyl H; and
methyl isobutyl methyl isobutyl.
[00238] Exemplary Amino Acid units include, but are not limited to, units of
formula
VII where: R20 is benzyl and R21 is -(CH2)4NH2; R20 is isopropyl and R21 is
-(CH2)4NH2, or R20 is isopropyl and R21 is -(CH2)3NHCONH2. Another exemplary
Amino
Acid unit is a unit of formula VIII wherein R20 is benzyl, R21 is benzyl, and
R22 is -
(CH2)4NH2.

63


CA 02754531 2011-09-02
WO 2010/111018 PCT/US2010/026429
[00239] Useful -W ,- units can be designed and optimized in their selectivity
for
enzymatic cleavage by a particular enzyme, for example, a tumor-associated
protease. In
one embodiment, a -W w - unit is that whose cleavage is catalyzed by cathepsin
B, C and
D, or a plasmin protease.
[00240] In one embodiment, -W ,- is a dipeptide, tripeptide, tetrapeptide or
pentapeptide. When R19, R20, R21, R22 or R23 is other than hydrogen, the
carbon atom to
which R19, R20, R21, R22 or R23 is attached is chiral.
[00241] Each carbon atom to which R19, R20, R21, R22 or R23 is attached is
independently in the (S) or (R) configuration.
[00242] In one aspect of the Amino Acid unit, the Amino Acid unit is valine-
citrulline
(vc or val-cit). In another aspect, the Amino Acid unit is phenylalanine-
lysine (i.e., fk).
In yet another aspect of the Amino Acid unit, the Amino Acid unit is N-
methylvaline-
citrulline. In yet another aspect, the Amino Acid unit is 5-aminovaleric acid,
homo
phenylalanine lysine, tetraisoquinolinecarboxylate lysine, cyclohexylalanine
lysine,
isonepecotic acid lysine, beta-alanine lysine, glycine serine valine glutamine
and
isonepecotic acid.

VIII.) The Spacer Unit
[00243] The Spacer unit (-Y-), when present, links an Amino Acid unit to the
Drug
unit when an Amino Acid unit is present. Alternately, the Spacer unit links
the Stretcher
unit to the Drug unit when the Amino Acid unit is absent. The Spacer unit also
links the
Drug unit to the Antibody unit when both the Amino Acid unit and Stretcher
unit are
absent.
[00244] Spacer units are of two general types: non self-immolative or self-
immolative.
A non self-immolative Spacer unit is one in which part or all of the Spacer
unit remains
bound to the Drug moiety after cleavage, particularly enzymatic, of an Amino
Acid unit
from the antibody-drug conjugate. Examples of a non self-immolative Spacer
unit
include, but are not limited to a (glycine-glycine) Spacer unit and a glycine
Spacer unit
(both depicted in Scheme 1) (infra). When a conjugate containing a glycine-
glycine
Spacer unit or a glycine Spacer unit undergoes enzymatic cleavage via an
enzyme (e.g., a
tumor-cell associated-protease, a cancer-cell-associated protease or a
lymphocyte-
associated protease), a glycine-glycine-Drug moiety or a glycine-Drug moiety
is cleaved

64


CA 02754531 2011-09-02
WO 2010/111018 PCT/US2010/026429
from L-Aa-Ww-. In one embodiment, an independent hydrolysis reaction takes
place
within the target cell, cleaving the glycine-Drug moiety bond and liberating
the Drug.
Scheme 1

L+Aa-Ww Gly-D, L+Aa-WW-Gly-G1y~D
enzymatic enzymatic
cleavage cleavage
Gly-D Gly-Gly-D
hydrolysis 1 hydrolysis 1

Drug Drug
[00245] In some embodiments, a non self-immolative Spacer unit (-Y-) is -Gly-.
In
some embodiments, a non self-immolative Spacer unit (-Y-) is -Gly-Gly-.
[00246] In one embodiment, a Drug-Linker conjugate is provided in which the
Spacer
unit is absent (y=0), or a pharmaceutically acceptable salt or solvate
thereof.
[00247] Alternatively, a conjugate containing a self-immolative Spacer unit
can release
-D. As used herein, the term "self-immolative Spacer" refers to a bifunctional
chemical
moiety that is capable of covalently linking together two spaced chemical
moieties into a
stable tripartite molecule. It will spontaneously separate from the second
chemical
moiety if its bond to the first moiety is cleaved.
[00248] In some embodiments, -Yy is a p-aminobenzyl alcohol (PAB) unit (see
Schemes 2 and 3) whose phenylene portion is substituted with Qm wherein Q is -
C1-C8
alkyl, -C1-C8 alkenyl, -C1-C8 alkynyl, -O-(C1-C8 alkyl), -O-(C1-C8 alkenyl), -
O-(C1-C8
alkynyl), -halogen, - nitro or -cyano; and m is an integer ranging from 0-4.
The alkyl,
alkenyl and alkynyl groups, whether alone or as part of another group, can be
optionally
substituted.
[00249] In some embodiments, -Y- is a PAB group that is linked to -W w - via
the
amino nitrogen atom of the PAB group, and connected directly to -D via a
carbonate,
carbamate or ether group. Without being bound by any particular theory or
mechanism,
Scheme 2 depicts a possible mechanism of Drug release of a PAB group which is
attached directly to -D via a carbamate or carbonate group as described by
Toki et al.,
2002, T. Org. Chem. 67:1866-1872.
Scheme 2


CA 02754531 2011-09-02
WO 2010/111018 PCT/US2010/026429
QM
L a WwiNH
O-CD
O P
enzymatic
cleavage
QM

O-CD
11
O
1,6-elimination
[00250] Drug
[00251] In Scheme 2, Q is -C1-C8 alkyl, -C1-C8 alkenyl, -Ci-C8 alkynyl, -O-(C1-
C8
alkyl), -O-(C1-C8 alkenyl), -O-(C1-C8 alkynyl), -halogen, -nitro or -cyano; m
is an integer
ranging from 0-4; and p ranges from 1 to about 20. The alkyl, alkenyl and
alkynyl
groups, whether alone or as part of another group, can be optionally
substituted.
[00252] Without being bound by any particular theory or mechanism, Scheme 3
depicts a possible mechanism of Drug release of a PAB group which is attached
directly
to -D via an ether or amine linkage, wherein D includes the oxygen or nitrogen
group that
is part of the Drug unit.
Scheme 3
66


CA 02754531 2011-09-02
WO 2010/111018 PCT/US2010/026429
QM
L Aa WW-NH -
D
enzymatic
cleavage
QM

NH2 \ /__J
D
1,6-elimination
P
+ Drug
[NHm1

[00253] In Scheme 3, Q is -C1-C8 alkyl, -C1-C8 alkenyl, -Ci-C8 alkynyl, -O-(C1-
C8
alkyl), -O-(C1-C8 alkenyl), -O-(C1-C8 alkynyl), -halogen, -nitro or -cyano; m
is an integer
ranging from 0-4; and p ranges from 1 to about 20. The alkyl, alkenyl and
alkynyl
groups, whether alone or as part of another group, can be optionally
substituted.
[00254] Other examples of self-immolative spacers include, but are not limited
to,
aromatic compounds that are electronically similar to the PAB group such as 2-
aminoimidazol- 5-methanol derivatives (Hay et al., 1999, Bioorg. Med. Chem.
Lett.
9:2237) and ortho or para-aminobenzylacetals. Spacers can be used that undergo
cyclization upon amide bond hydrolysis, such as substituted and unsubstituted
4-
aminobutyric acid amides (Rodrigues et al., 1995, Chemistry Biology 2:223),
appropriately substituted bicyclo[2.2.1] and bicyclo[2.2.2] ring systems
(Storm et al.,
1972, T. Amer. Chem. Soc. 94:5815) and 2-aminophenylpropionic acid amides
(Amsberry
et al., 1990, T. Org. Chem. 55:5867). Elimination of amine-containing drugs
that are
substituted at the a-position of glycine (Kingsbury et al., 1984, T. Med.
Chem. 27:1447)
are also examples of self-immolative spacers.

67


CA 02754531 2011-09-02
WO 2010/111018 PCT/US2010/026429
[00255] In one embodiment, the Spacer unit is a branched bis(hydroxymethyl) -
styrene
(BHMS) unit as depicted in Scheme 4, which can be used to incorporate and
release
multiple drugs.
Scheme 4

Om CH2(O(C(O)))n-D
L Aa WW-NH CH2(O(C(O)))n D
0
P
enzymatic
cleavage
[00256] 2 drugs
[00257] In Scheme 4, Q is -Ci-C8 alkyl, -Ct-C8 alkenyl, -Ct-C8 alkynyl, -O-(CI-
C8
alkyl), -O-(CI-C8 alkenyl), -O-(CI-C8 alkynyl), -halogen, -nitro or -cyano; m
is an integer
ranging from 0-4; n is 0 or 1; and p ranges raging from 1 to about 20. The
alkyl, alkenyl
and alkynyl groups, whether alone or as part of another group, can be
optionally
substituted.
[00258] In some embodiments, the -D moieties are the same. In yet another
embodiment, the -D moieties are different.
[00259] In one aspect, Spacer units (-Yy) are represented by Formulas (X)-
(XII):
H Qm
N

OJT"
[00260] 0 x
[00261] wherein Q is -Ci-C8 alkyl, -Ci-C8 alkenyl, -Ct-C8 alkynyl, -O-(CI-C8
alkyl), -
O-(CI-C8 alkenyl), -O-(CI-C8 alkynyl), -halogen, -nitro or -cyano; and m is an
integer
ranging from 0-4. The alkyl, alkenyl and alkynyl groups, whether alone or as
part of
another group, can be optionally substituted.

[00262] ~xi
and

-N HCH2C(O)-N HCH2C(O)-
[00263] XII.
68


CA 02754531 2011-09-02
WO 2010/111018 PCT/US2010/026429
[00264] Embodiments of the Formula I and II comprising antibody-drug conjugate
compounds can include:

0
N Ww-Y,-D
L o
p
[00265]
[00266] wherein w and y are each 0, 1 or 2, and,
0
N D
L
0 S 0
p
[00267]
[00268] wherein w and y are each 0,
0
0 O~D
H L Aa-HN
:fy N ''A N
H
O

NH
O=<
[00269] NH2
O
L S 0
N N
H H-Yy - D
0 o P
NH
O(
[00270] NH2
and
69


CA 02754531 2011-09-02
WO 2010/111018 PCT/US2010/026429
O
L S O H O I/ O D
N N N,,~,kN
O O H P
NH
O=<
NH2
IX.) The Drug Unit

[00271] The Drug moiety (D) can be any cytotoxic, cytostatic or
immunomodulatory
(e.g., immunosuppressive) or drug. D is a Drug unit (moiety) having an atom
that can
form a bond with the Spacer unit, with the Amino Acid unit, with the Stretcher
unit or
with the Antibody unit. In some embodiments, the Drug unit D has a nitrogen
atom that
can form a bond with the Spacer unit. As used herein, the terms "Drug unit"
and "Drug
moiety" are synonymous and used interchangeably.
[00272] Useful classes of cytotoxic or immunomodulatory agents include, for
example,
antitubulin agents, DNA minor groove binders, DNA replication inhibitors, and
alkylating agents.
[00273] In some embodiments, the Drug is an auristatin, such as auristatin E
(also
known in the art as a derivative of dolastatin-10) or a derivative thereof.
The auristatin
can be, for example, an ester formed between auristatin E and a keto acid. For
example,
auristatin E can be reacted with paraacetyl benzoic acid or benzoylvaleric
acid to produce
AEB and AEVB, respectively. Other typical auristatins include AFP, MMAF, and
MMAE. The synthesis and structure of exemplary auristatins are described in
U.S. Patent
Application Publication Nos. 2003-0083263, 2005-0238649 and 2005-0009751;
International Patent Publication No. WO 04/010957, International Patent
Publication No.
WO 02/088172, and U.S. Patent Nos. 6,323,315; 6,239,104; 6,034,065; 5,780,588;
5,665,860; 5,663,149; 5,635,483; 5,599,902; 5,554,725; 5,530,097; 5,521,284;
5,504,191;
5,410,024; 5,138,036; 5,076,973; 4,986,988; 4,978,744; 4,879,278; 4,816,444;
and
4,486,414, each of which is incorporated by reference herein in its entirety
and for all
purposes.



CA 02754531 2011-09-02
WO 2010/111018 PCT/US2010/026429
[00274] Auristatins have been shown to interfere with microtubule dynamics and
nuclear and cellular division and have anticancer activity. Auristatins bind
tubulin and
can exert a cytotoxic or cytostatic effect on a 24P4C12-expressing cell. There
are a
number of different assays, known in the art, which can be used for
determining whether
an auristatin or resultant antibody-drug conjugate exerts a cytostatic or
cytotoxic effect on
a desired cell line.
[00275] Methods for determining whether a compound binds tubulin are known in
the
art. See, for example, Muller et al., Anal. Chem 2006, 78, 4390-4397; Hamel et
al.,
Molecular Pharmacology, 1995 47: 965-976; and Hamel et al., The Journal of
Biological
Chemistry, 1990 265:28, 17141-17149. For purposes of the present invention,
the relative
affinity of a compound to tubulin can be determined. Some preferred
auristatins of the
present invention bind tubulin with an affinity ranging from 10 fold lower
(weaker
affinity) than the binding affinity of MMAE to tubulin to 10 fold, 20 fold or
even 100 fold
higher (higher affinity) than the binding affinity of MMAE to tublin.
[00276]
In some embodiments, -D is an auristatin of the formula DE or DF:
R3 O R7
R9 R25
N I
N N N N
R24
R2 O R4 R R6 R8 O
R8 O R26
DE

R3 O R7
R9 O
N I
N N N N ZR11
R2 O R4 5 R6 R8 O
$
R1o
DF

or a pharmaceutically acceptable salt or solvate form thereof;
wherein, independently at each location:
the wavy line indicates a bond;

71


CA 02754531 2011-09-02
WO 2010/111018 PCT/US2010/026429
R 2 is -Ci-C20 alkyl, -C2-C20 alkenyl, or -C2-C20 alkynyl;
R3 is -H, -Ci-C20 alkyl, -C2-C20 alkenyl, -C2-C20 alkynyl, -carbocycle, -Ci-
C20
alkylene (carbocycle), -C2-C20 alkenylene(carbocycle), -C2-C20
alkynylene(carbocycle), -
aryl, -CI-C20 alkylene(aryl), -C2-C20 alkenylene(aryl), -C2-C20
alkynylene(aryl),
heterocycle, -Ci-C20 alkylene(heterocycle), -C2-C20 alkenylene(heterocycle),
or -C2-C20
alkynylene(heterocycle);
R4 is -H, -Ci-C20 alkyl, -C2-C20 alkenyl, -C2-C20 alkynyl, carbocycle, -Ci-C20
alkylene (carbocycle), -C2-C20 alkenylene(carbocycle), -C2-C20
alkynylene(carbocycle),
aryl, -CI-C20 alkylene(aryl), -C2-C20 alkenylene(aryl), -C2-C20
alkynylene(aryl), -
heterocycle, -Ci-C20 alkylene(heterocycle), -C2-C20 alkenylene(heterocycle),
or -C2-C20
alkynylene(heterocycle);
R5 is -H or -C1-C8 alkyl;
or R4 and R5 jointly form a carbocyclic ring and have the
formula -(CRaRb)s- wherein Ra and Rb are independently -H, -Ci-C20 alkyl, -C2-
C20
alkenyl, -C2-C2o alkynyl, or -carbocycle and s is 2, 3, 4, 5 or 6,
R6 is -H, -Ci-C20 alkyl, -C2-C20 alkenyl, or -C2-C2o alkynyl;
R7 is -H, -Ci-C20 alkyl, -C2-C20 alkenyl, -C2-C20 alkynyl, carbocycle, -Ci-C20
alkylene (carbocycle), -C2-C20 alkenylene(carbocycle), -C2-C20
alkynylene(carbocycle), -
aryl, -Ci-C20 alkylene(aryl), -C2-C20 alkenylene(aryl), -C2-C20
alkynylene(aryl),
heterocycle, -Ci-C20 alkylene(heterocycle), -C2-C2o alkenylene(heterocycle),
or -C2-C20
alkynylene(heterocycle);
each R8 is independently -H, -OH, -Ci-C20 alkyl, -C2-C2o alkenyl, -C2-C20
alkynyl, -O-(C1-C20 alkyl), -O-(C2-C20 alkenyl), -O-(C1-C20 alkynyl), or -
carbocycle;
R9 is -H, -Ci-C20 alkyl, -C2-C20 alkenyl, or -C2-C20 alkynyl;
R24 is -aryl, -heterocycle, or -carbocycle;
R25 is -H, CI-C20 alkyl, -C2-C20 alkenyl, -C2-C20 alkynyl, -carbocycle, -O-(Ci-

C20 alkyl),
-O-(C2-C20 alkenyl), -O-(C2-C20 alkynyl), or OR'8 wherein Rig is -H, a
hydroxyl
protecting group, or a direct bond where OR'8 represents =O;
R26 is -H, -CI-C20 alkyl, -C2-C20 alkenyl, or -C2-C20 alkynyl, -aryl, -
heterocycle,
or

-carbocycle;

72


CA 02754531 2011-09-02
WO 2010/111018 PCT/US2010/026429
R10 is -aryl or -heterocycle;
Z is -0, -S, -NH, or -NR12, wherein R'2 is -Ci-C20 alkyl, -C2-C20 alkenyl, or -
C2-
C20 alkynyl;
R" is -H, -C1-C2o alkyl, --C2-C20 alkenyl, -C2-C20 alkynyl, -aryl, -
heterocycle, -
(Ri30)mRio or -(R130)m CH(Ris)2;;

m is an integer ranging from 1-1000;
R13 is -C2-C20 alkylene, -C2-C20 alkenylene, or -C2-C20 alkynylene;
R14 is -H, -Ci-C20 alkyl, -C2-C20 alkenyl, or -C2-C20 alkynyl;

each occurrence of R15 is independently -H, -000H, -(CH2)n-N(R16)2, -(CH2)n-
S0311, -(CH2)n-SO3-CI-C20 alkyl, -(CH2)n-SO3-C2-C20 alkenyl, or -(CH2)n-SO3-C2-
C20
alkynyl;
each occurrence of R16 is independently -H, -Ci-C20 alkyl, -C2-C2o alkenyl, -
C2-
C20 alkynyl or -(CH2)n-000H; and

n is an integer ranging from 0 to 6;
wherein said alkyl, alkenyl, alkynyl, alkylene, alkenylene, alkynyklene, aryl,
carbocyle, and heterocycle radicals, whether alone or as part of another
group, are
optionally substituted.
Auristatins of the formula DE include those wherein said alkyl, alkenyl,
alkynyl,
alkylene, alkenylene, alkynyklene, aryl, carbocyle, and heterocycle radicals
are
unsubstituted.
Auristatins of the formula DE include those wherein the groups of R2, R3, R4,
Rs,
R6, R7, R8, and R9 are unsubstituted and the groups of R19, R2 and R2' are
optionally
substituted as described herein.
Auristatins of the formula DE include those wherein
R2 is CI-C8 alkyl;
R3, R4 and R7 are independently selected from -H, -Ci-C20 alkyl, -C2-C20
alkenyl, -C2-C2o alkynyl, monocyclic C3-C6 carbocycle, -Ci-C2o
alkylene(monocyclic C3-
C6 carbocycle), -C2-C20 alkenylene(monocyclic C3-C6 carbocycle), -C2-C20
alkynylene(monocyclic C3-C6 carbocycle), C6-C10 aryl, -Ci-C20 alkylene(C6-Cio
aryl), -
C2-C20 alkenylene(C6-Cio aryl), -C2-C20 alkynylene(C6-Cio aryl), heterocycle, -
Ci-C20
alkylene(heterocycle), -C2-C2o alkenylene(heterocycle), or -C2-C20

73


CA 02754531 2011-09-02
WO 2010/111018 PCT/US2010/026429
alkynylene(heterocycle); wherein said alkyl, alkenyl, alkynyl, alkylene,
alkenylene,
alkynylene, carbocycle, aryl and heterocycle radicals are optionally
substituted;
R5 is -H;
R6 is -C1-C8 alkyl;
each R8 is independently selected from -OH, -O-(C1-C20 alkyl), -O-(C2-C20
alkenyl), or
-O-(C2-C20 alkynyl) wherein said alkyl, alkenyl, and alkynyl radicals are
optionally
substituted;
R9 is -H or -CI-C8 alkyl;
R24 is optionally substituted -phenyl;
R25 is -OR'8; wherein Rig is H, a hydroxyl protecting group, or a direct bond
where OR'8 represents =O;
R26 is selected from -H, -Ci-C20 alkyl, -C2-C20 alkenyl, -C2-C20 alkynyl, or -
carbocycle; wherein said alkyl, alkenyl, alkynyl and carbocycle radicals are
optionally
substituted; or a pharmaceutically acceptable salt or solvate form thereof.
Auristatins of the formula DE include those wherein
R2 is methyl;
R3 is -H, -C1-C8 alkyl, -C2-C8 alkenyl, or C2-C8 alkynyl, wherein said alkyl,
alkenyl and alkynyl radicals are optionally substituted;
R4 is -H, -C1-C8 alkyl, -C2-C8 alkenyl, -C2-C8 alkynyl, monocyclic C3-C6
carbocycle, -C6-C10 aryl, -CI-C8 alkylene(C6-Ci0 aryl), -C2-C8 alkenylene(C6-
Ci0 aryl), -
C2-C8 alkynylene(C6-Ci0 aryl), -CI-C8 alkylene (monocyclic C3-C6 carbocycle), -
C2-C8
alkenylene (monocyclic C3-C6 carbocycle), -C2-C8 alkynylene(monocyclic C3-C6
carbocycle); wherein said alkyl, alkenyl, alkynyl, alkylene, alkenylene,
alkynylene, aryl
and carbocycle radicals whether alone or as part of another group are
optionally
substituted;
R5 is -H;
R6 is methyl;
R7 is -C1-C8 alkyl, -C2-C8 alkenyl or -C2-C8 alkynyl;
each R8 is methoxy;

R9 is -H or -CI-C8 alkyl;
R24 is -phenyl;

74


CA 02754531 2011-09-02
WO 2010/111018 PCT/US2010/026429
R25 is -OR"; wherein Rig is H, a hydroxyl protecting group, or a direct bond
where OR'8 represents =O;
R26 is methyl;
or a pharmaceutically acceptable salt form thereof.
Auristatins of the formula DE include those wherein:
R2 is methyl; R3 is -H or -C1-C3 alkyl; R4 is -C1-C5 alkyl; R5 is -H; R6 is
methyl; R7 is
isopropyl or sec-butyl; R8 is methoxy; R9 is -H or -C1-C8 alkyl; R24 is
phenyl; R25 is -
OR'8; wherein Rig is -H, a hydroxyl protecting group, or a direct bond where
OR'8
represents =O; and R26 is methyl; or a pharmaceutically acceptable salt or
solvate form
thereof.
Auristatins of the formula DE include those wherein:
R2 is methyl or CI-C3 alkyl,
R3 is -H or -C,-C3 alkyl;
R4 is -C,-C5 alkyl;
R5 is H;
R6 is C1-C3 alkyl;
R7 is -C,-C5 alkyl;
R8 is -C,-C3 alkoxy;
R9 is -H or -C,-C8 alkyl;
R24 is phenyl;

R25 is -OR'8; wherein Rig is -H, a hydroxyl protecting group, or a direct bond
where OR'8 represents =O; and
R26 is -C,-C3 alkyl;
or a pharmaceutically acceptable salt form thereof.
Auristatins of the formula DF include those wherein
R2 is methyl;
R3, R4, and R7 are independently selected from -H, -Ci-C20 alkyl, -C2-C20
alkenyl, -C2-C20 alkynyl, monocyclic C3-C6 carbocycle, -C,-CM
alkylene(monocyclic C3-
C6 carbocycle), -C2-C20 alkenylene(monocyclic C3-C6 carbocycle), -C2-C20
alkynylene(monocyclic C3-C6 carbocycle), -C6-C10 aryl, -Ci-C20 alkylene(C6-C10
aryl), -
C2-C20 alkenylene(C6-C10 aryl), -C2-C20 alkynylene(C6-C10 aryl), heterocycle, -
Ci-C20
alkylene(heterocycle), -C2-C20 alkenylene(heterocycle), or -C2-C20



CA 02754531 2011-09-02
WO 2010/111018 PCT/US2010/026429
alkynylene(heterocycle); wherein said alkyl, alkenyl, alkynyl, alkylene,
alkenylene,
alkynylene, carbocycle, aryl and heterocycle radicals whether alone or as part
of another
group are optionally substituted;
R5 is -H;
R6 is methyl;
each R8 is methoxy;
R9 is -H, -Ci-C20 alkyl, -C2-C20 alkenyl, or -C2-C20 alkynyl; wherein said
alkyl,
alkenyl and alkynyl radical are optionally substituted;
R10 is optionally substituted aryl or optionally substituted heterocycle;
Z is -0-, -S-, -NH-, or -NR12, wherein R'2 is -Ci-C20 alkyl, -C2-C20 alkenyl,
or -
C2-C20 alkynyl, each of which is optionally substituted;
R" is -H, -CI-C20 alkyl, -C2-C20 alkenyl, -C2-C20 alkynyl, -aryl, -
heterocycle, -
(R13O)m R14, or -(R13O)m CH(R'5)2, wherein said alkyl, alkenyl, alkynyl, aryl
and
heterocycle radicals are optionally substituted;
m is an integer ranging from 1-1000 or m = 0;
R13 is -C2-C20 alkylene, -C2-C20 alkenylene, or -C2-C20 alkynylene, each of
which is optionally substituted;
R14 is -H, -Ci-C20 alkyl, -C2-C20 alkenyl, or -C2-C20 alkynyl wherein said
alkyl,
alkenyl and alkynyl radicals are optionally substituted;

each occurrence of R15 is independently -H, -000H, -(CH2)n-N(R16)2, -(CH2)n-
S0314, -(CH2)n-SO3-C1-C20 alkyl, -(CH2)n-SO3-C2-C20 alkenyl, or -(CH2)n-SO3-C2-
C20
alkynyl wherein said alkyl, alkenyl and alkynyl radicals are optionally
substituted;
each occurrence of R16 is independently -H, -Ci-C20 alkyl, -C2-C20 alkenyl, -
C2-
C20 alkynyl or -(CH2)n-000H wherein said alkyl, alkenyl and alkynyl radicals
are
optionally substituted;
n is an integer ranging from 0 to 6;
or a pharmaceutically acceptable salt thereof.
In certain of these embodiments, R10 is optionally substituted phenyl.
Auristatins of the formula DF include those wherein the groups of R2, R3, R4,
R5,
R6, R7, R8, and R9 are unsubstituted and the groups of R10 and R" are as
described herein.
76


CA 02754531 2011-09-02
WO 2010/111018 PCT/US2010/026429
Auristatins of the formula DF include those wherein said alkyl, alkenyl,
alkynyl,
alkylene, alkenylene, alkynyklene, aryl, carbocyle, and heterocycle radicals
are
unsubstituted
Auristatins of the formula DF include those wherein
R2 is -Ci-C3 alkyl; R3 is -H or -Ci-C3 alkyl; R4 is -Ci-C5 alkyl; R 5 is -H;
R6 is -
CI-C3 alkyl; R7 is -C1-C5 alkyl; R8 is -C1-C3 alkoxy; R9 is -H or -Ci-C8
alkyl; R10 is
optionally substituted phenyl; Z is -0-, -S-, or -NH-; R" is as defined
herein; or a
pharmaceutically acceptable salt thereof.
Auristatins of the formula DF include those wherein
R2 is methyl; R3 is -H or -Ci-C3 alkyl; R4 is -Ci-C5 alkyl; R 5 is -H; R6 is
methyl;
R7 is isopropyl or sec-butyl; R8 is methoxy; R9 is -H or -C1-C8 alkyl; R10 is
optionally
substituted phenyl; Z is -0-, -S-, or -NH-; and R" is as defined herein; or a
pharmaceutically acceptable salt thereof.
Auristatins of the formula DF include those wherein
R2 is methyl; R3 is -H or -Ci-C3 alkyl; R4 is -Ci-C5 alkyl; R 5 is -H; R6 is
methyl;
R7 is isopropyl or sec-butyl; R8 is methoxy; R9 is -H or Ci-C8 alkyl; R10 is
phenyl; and Z
is -0- or -NH- and R" is as defined herein, preferably hydrogen; or a
pharmaceutically
acceptable salt form thereof.
Auristatins of the formula DF include those wherein
R2 is -Ci-C3 alkyl; R3 is -H or -Ci-C3 alkyl; R4 is -Ci-C5 alkyl; R 5 is -H;
R6 is -
CI-C3 alkyl; R7 is -C1-C5 alkyl; R8 is -Ci-C3 alkoxy; R9 is -H or -C1-C8
alkyl; R10 is
phenyl; and Z is -0- or -NH- and R" is as defined herein, preferably hydrogen;
or a
pharmaceutically acceptable salt form thereof.
Auristatins of the formula DE or DF include those wherein R3, R4 and R7 are
independently isopropyl or sec-butyl and R5 is -H. In an exemplary embodiment,
R3 and
R4 are each isopropyl, R5 is H, and R7 is sec-butyl. The remainder of the
substituents are
as defined herein.
Auristatins of the formula DE or DF include those wherein R2 and R6 are each
methyl, and R9 is H. The remainder of the substituents are as defined herein.
Auristatins of the formula DE or DF include those wherein each occurrence of
R8
is -OCH3. The remainder of the substituents are as defined herein.

77


CA 02754531 2011-09-02
WO 2010/111018 PCT/US2010/026429
Auristatins of the formula DE or DF include those wherein R3 and R4 are each
isopropyl, R2 and R6 are each methyl, R5 is H, R7 is sec-butyl, each
occurrence of R8 is -
OCH3, and R9 is H. The remainder of the substituents are as defined herein.
Auristatins of the formula DF include those wherein Z is -0- or -NH-. The
remainder of the substituents are as defined herein.
Auristatins of the formula DF include those wherein R10 is aryl. The remainder
of
the substituents are as defined herein.
Auristatins of the formula DF include those wherein R10 is -phenyl. The
remainder of the substituents are as defined herein.
Auristatins of the formula DF include those wherein Z is -0-, and R11 is H,
methyl
or t-butyl. The remainder of the substituents are as defined herein.
Auristatins of the formula DF include those wherein, when Z is -NH-, R" is -
R130 õ-CH(R15 is i i6 i6
( ) )2, wherein R15 is -(CH2)n-N(R )2, and R is -Ci-Cg alkyl or -(CH2)n-
COOH. The remainder of the substituents are as defined herein.
Auristatins of the formula DF include those wherein when Z is -NH-, R" is -
(R13O)m CH(R'5)2, wherein R'5 is -(CH2)n-SO3H. The remainder of the
substituents are
as defined herein.
In preferred embodiments, when D is an auristatin of formula DE, w is an
integer
ranging from 1 to 12, preferably 2 to 12, y is 1 or 2, and a is preferably 1.
In some embodiments, wheren D is is an auristatin of formula DF, a is 1 and w
and
y are 0.
Illustrative Drug units (-D) include the drug units having the following
structures:
O CH3
N N N N NH
0 OCH3O OCH3O
O OH
78


CA 02754531 2011-09-02
WO 2010/111018 PCT/US2010/026429
O CH3 OH
N N N N NH o

0 ~ OCH3O OCH3O

O OH
N N
O O O O O

O H
\s H
S N NN N N
I Y
0 I O O 0 O O OH
N N
N H O\
O H O OCH3O OCH3O
~ry

O
N N N N
0 O_ 0
O'-, 0 0 O
79


CA 02754531 2011-09-02
WO 2010/111018 PCT/US2010/026429
O
H
N'' N N N
O O_ 0
Ol-~ OO NH /
H
O po
N N N
O OCH3 0 H
OCH3 O 0
O
/' N
' N N N
N
O O_ O
O1-1 O O NH
H
O
H
N N N N
O O'~, O O1-1 O
O O
HOOCI---,,N,---.,COOH
H O
N N N N
O I O- O O O
O NH
H
SO3H


CA 02754531 2011-09-02
WO 2010/111018 PCT/US2010/026429
O

N
:~N
N N N N
1~~
O O- O O1-1 O
O NH /
HOOC

COOH , and
o

N i N N
O O
O1_~ O O NH
NH2
or pharmaceutically acceptable salts or solvates thereof.
In one aspect, hydrophilic groups, such as but not limited to triethylene
glycol
esters (TEG) can be attached to the Drug Unit at R". Without being bound by
theory, the
hydrophilic groups assist in the internalization and non-agglomeration of the
Drug Unit.
In some embodiments, the Drug unit is not TZT-1027. In some embodiments, the
Drug unit is not auristatin E, dolastatin 10, or auristatin PE.
Exemplary antibody-drug conjugate compounds have the following structures
wherein "L" or "mAb-s-" represents an 24P4C12 MAb designated Ha5-1(5)2.1 set
forth
herein:

O 0 ON N O N N N \
N / 0 OCH30 OCH30I
L Val-Cit-HN I 0 \%
OH
0
P
81


CA 02754531 2011-09-02
WO 2010/111018 PCT/US2010/026429
II OH
O O Oi N O i N N
N V I- v p 0CH30
Cit-N I
OCH30
L
H
O
P
H pC CH3 H

H j\ O NN N'~ ^ ~I ~N
/ OCHO OCH30
N I O
N N N 0=~`
mAb S O H 0 H OH
P
NH
O=(
NH2
L-MC-vc-PAB-MMAF
or
Error! Objects cannot be created from editing field codes.
L-MC-vc-PAB-MMAE.

or

O 3C ! CH3
S N NNE' N~~^~f~NH
_~,, ~y
mAb i
O O OCH30 OCH30
O OH
L-MC-MMAF
or pharmaceutically acceptable salt thereof.
In some embodiments, the Drug Unit is a calicheamicin, camptothecin, a
maytansinoid, or an anthracycline. In some embodiments the drug is a taxane, a
topoisomerase inhibitor, a vinca alkaloid, or the like.
In some typical embodiments, suitable cytotoxic agents include, for example,
DNA minor groove binders (e.g., enediynes and lexitropsins, a CBI compound;
see also
U.S. Patent No. 6,130,237), duocarmycins, taxanes (e.g., paclitaxel and
docetaxel),
puromycins, and vinca alkaloids. Other cytotoxic agents include, for example,
CC-1065,
SN-38, topotecan, morpholino-doxorubicin, rhizoxin, cyanomorpholino-
doxorubicin,

82


CA 02754531 2011-09-02
WO 2010/111018 PCT/US2010/026429
echinomycin, combretastatin, netropsin, epothilone A and B, estramustine,
cryptophysins,
cemadotin, maytansinoids, discodermolide, eleutherobin, and mitoxantrone.
In some embodiments, the Drug is an anti-tubulin agent. Examples of anti-
tubulin
agents include, auristatins, taxanes (e.g., Taxol (paclitaxel), Taxotere
(docetaxel)),
T67 (Tularik) and vinca alkyloids (e.g., vincristine, vinblastine, vindesine,
and
vinorelbine). Other antitubulin agents include, for example, baccatin
derivatives, taxane
analogs (e.g., epothilone A and B), nocodazole, colchicine and colcimid,
estramustine,
cryptophycins, cemadotin, maytansinoids, combretastatins, discodermolide, and
eleutherobin.
In certain embodiments, the cytotoxic agent is a maytansinoid, another group
of
anti-tubulin agents. For example, in specific embodiments, the maytansinoid is
maytansine or DM-1 (ImmunoGen, Inc.; see also Chari et al., 1992, Cancer Res.
52:127-
131).
In certain embodiments, the cytotoxic or cytostatic agent is a dolastatin. In
certain
embodiments, the cytotoxic or cytostatic agent is of the auristatin class.
Thus, in a
specific embodiment, the cytotoxic or cytostatic agent is MMAE (Formula XI).
In
another specific embodiment, the cytotoxic or cytostatic agent is AFP (Formula
XVI).
H3C CH3 H3C
O CH3 HO
H CH3
N'=.
HN N N
CH3 O CH3 OCH3 0 H CH3
H3C CH3 OCH3 0
(XI)
In certain embodiments, the cytotoxic or cytostatic agent is a compound of
formulas XII-XXI or pharmaceutically acceptable salt thereof:

83


CA 02754531 2011-09-02
WO 2010/111018 PCT/US2010/026429
NHZ
O
H
N N
N
11O OCH3 O H
OCH3 0

(XII)
O
H
N''-,
N
~ I N
O OCH3 O H
OCH3 0
(XIII)
H3C CH3 H3C
O CH3 H CH3

HN N s
N
CH3 0 CH3 OCH3 0 H
CH3 OCH3 0 N
CH3

(XIV)
84


CA 02754531 2011-09-02
WO 2010/111018 PCT/US2010/026429
O H
H ZN

N 'NO \
N """'a
0 OCH3 O H
OCH3 0 /
(XV)

H3C CH3 H3C
O CH3 NH2
O
H CH3
H3Cl, N,, H
N N N N,,, <::f
N
CH3 0 CHOCH3 0 H

H3C CH3 OCH3 0 (XVI)

H3C CH3 H3C
O CH3 H CH3

HI i S
H N
CH3 0 CH3 0 H
H3C CH3 OCH3 0
N x
(XVII)

NH2
H
O PN
-r~ Ijr N N N
N
O OCH3 O H
OCH3 O 0
(XVIII)



CA 02754531 2011-09-02
WO 2010/111018 PCT/US2010/026429
H
HN N N N
Y N

/
O O O O O O OH

(XVIV)

0

YO
O O
H
N N N
Y N
N
O OCH3 O H
OCH3 0

(XX)
0

O
O O
H
N
Y N N
N
O OCH3 O H
OCH3 0

(XXI)
X.) Drug Loading
[00277] Drug loading is represented by p and is the average number of Drug
moieties
per antibody in a molecule. Drug loading may range from 1 to 20 drug moieties
(D) per
antibody. ADCs of the invention include collections of antibodies conjugated
with a
range of drug moieties, from 1 to 20. The average number of drug moieties per
antibody
in preparations of ADC from conjugation reactions may be characterized by
conventional
means such as mass spectroscopy and, ELISA assay. The quantitative
distribution of

86


CA 02754531 2011-09-02
WO 2010/111018 PCT/US2010/026429
ADC in terms of p may also be determined. In some instances, separation,
purification,
and characterization of homogeneous ADC where p is a certain value from ADC
with
other drug loadings may be achieved by means such as electrophoresis.
[00278] For some antibody-drug conjugates, p may be limited by the number of
attachment sites on the antibody. For example, where the attachment is a
cysteine thiol,
as in the exemplary embodiments above, an antibody may have only one or
several
cysteine thiol groups, or may have only one or several sufficiently reactive
thiol groups
through which a linker may be attached. In certain embodiments, higher drug
loading,
e.g. p >5, may cause aggregation, insolubility, toxicity, or loss of cellular
permeability of
certain antibody-drug conjugates. In certain embodiments, the drug loading for
an ADC
of the invention ranges from 1 to about 8; from about 2 to about 6; from about
3 to about
5; from about 3 to about 4; from about 3.1 to about 3.9; from about 3.2 to
about 3.8; from
about 3.2 to about 3.7; from about 3.2 to about 3.6; from about 3.3 to about
3.8; or from
about 3.3 to about 3.7. Indeed, it has been shown that for certain ADCs, the
optimal ratio
of drug moieties per antibody may be less than 8, and may be about 2 to about
5. See US
2005-0238649 Al (herein incorporated by reference in its entirety).
[00279] In certain embodiments, fewer than the theoretical maximum of drug
moieties
are conjugated to an antibody during a conjugation reaction. An antibody may
contain,
for example, lysine residues that do not react with the drug-linker
intermediate or linker
reagent, as discussed below. Generally, antibodies do not contain many free
and reactive
cysteine thiol groups which may be linked to a drug moiety; indeed most
cysteine thiol
residues in antibodies exist as disulfide bridges. In certain embodiments, an
antibody
may be reduced with a reducing agent such as dithiothreitol (DTT) or
tricarbonylethylphosphine (TCEP), under partial or total reducing conditions,
to generate
reactive cysteine thiol groups. In certain embodiments, an antibody is
subjected to
denaturing conditions to reveal reactive nucleophilic groups such as lysine or
cysteine.
[00280] The loading (drug/antibody ratio) of an ADC may be controlled in
different
ways, e.g., by: (i) limiting the molar excess of drug-linker intermediate or
linker reagent
relative to antibody, (ii) limiting the conjugation reaction time or
temperature, (iii) partial
or limiting reductive conditions for cysteine thiol modification, (iv)
engineering by
recombinant techniques the amino acid sequence of the antibody such that the
number
and position of cysteine residues is modified for control of the number and/or
position of
87


CA 02754531 2011-09-02
WO 2010/111018 PCT/US2010/026429
linker-drug attachements (such as thioMab or thioFab prepared as disclosed
herein and in
W02006/034488 (herein incorporated by reference in its entirety)).
[00281] It is to be understood that where more than one nucleophilic group
reacts with
a drug-linker intermediate or linker reagent followed by drug moiety reagent,
then the
resulting product is a mixture of ADC compounds with a distribution of one or
more drug
moieties attached to an antibody. The average number of drugs per antibody may
be
calculated from the mixture by a dual ELISA antibody assay, which is specific
for
antibody and specific for the drug. Individual ADC molecules may be identified
in the
mixture by mass spectroscopy and separated by HPLC, e.g. hydrophobic
interaction
chromatography (see, e.g., Hamblett, K.J., et al. "Effect of drug loading on
the
pharmacology, pharmacokinetics, and toxicity of an anti-CD30 antibody-drug
conjugate,"
Abstract No. 624, American Association for Cancer Research, 2004 Annual
Meeting,
March 27-31, 2004, Proceedings of the AACR, Volume 45, March 2004; Alley,
S.C., et
al. "Controlling the location of drug attachment in antibody-drug conjugates,"
Abstract
No. 627, American Association for Cancer Research, 2004 Annual Meeting, March
27-
31, 2004, Proceedings of the AACR, Volume 45, March 2004). In certain
embodiments,
a homogeneous ADC with a single loading value may be isolated from the
conjugation
mixture by electrophoresis or chromatography.

XI.) Methods of Determining Cytotoxic effect of ADCs
[00282] Methods of determining whether a Drug or Antibody-Drug conjugate
exerts a
cytostatic and/or cytotoxic effect on a cell are known. Generally, the
cytotoxic or
cytostatic activity of a Antibody Drug conjugate can be measured by: exposing
mammalian cells expressing a target protein of the Antibody Drug conjugate in
a cell
culture medium; culturing the cells for a period from about 6 hours to about 5
days; and
measuring cell viability. Cell-based in vitro assays can be used to measure
viability
(proliferation), cytotoxicity, and induction of apoptosis (caspase activation)
of the
Antibody Drug conjugate.
[00283] For determining whether a Antibody Drug conjugate exerts a cytostatic
effect,
a thymidine incorporation assay may be used. For example, cancer cells
expressing a
target antigen at a density of 5,000 cells/well of a 96-well plated can be
cultured for a 72-
hour period and exposed to 0.5 Ci of 3H-thymidine during the final 8 hours of
the 72-

88


CA 02754531 2011-09-02
WO 2010/111018 PCT/US2010/026429
hour period. The incorporation of 3H-thymidine into cells of the culture is
measured in
the presence and absence of the Antibody Drug conjugate.
[00284] For determining cytotoxicity, necrosis or apoptosis (programmed cell
death)
can be measured. Necrosis is typically accompanied by increased permeability
of the
plasma membrane; swelling of the cell, and rupture of the plasma membrane.
Apoptosis
is typically characterized by membrane blebbing, condensation of cytoplasm,
and the
activation of endogenous endonucleases. Determination of any of these effects
on cancer
cells indicates that a Antibody Drug conjugate is useful in the treatment of
cancers.
[00285] Cell viability can be measured by determining in a cell the uptake of
a dye
such as neutral red, trypan blue, or ALAMARTM blue (see, e.g., Page et al.,
1993, Intl. T.
Oncology 3:473-476). In such an assay, the cells are incubated in media
containing the
dye, the cells are washed, and the remaining dye, reflecting cellular uptake
of the dye, is
measured spectrophotometrically. The protein-binding dye sulforhodamine B
(SRB) can
also be used to measure cytoxicity (Skehan et al., 1990, T. Natl. Cancer Inst.
82:1107-12).
[00286] Alternatively, a tetrazolium salt, such as MTT, is used in a
quantitative
colorimetric assay for mammalian cell survival and proliferation by detecting
living, but
not dead, cells (see, e.g., Mosmann, 1983, T. Immunol. Methods 65:55-63).
[00287] Apoptosis can be quantitated by measuring, for example, DNA
fragmentation.
Commercial photometric methods for the quantitative in vitro determination of
DNA
fragmentation are available. Examples of such assays, including TUNEL (which
detects
incorporation of labeled nucleotides in fragmented DNA) and ELISA-based
assays, are
described in Biochemica, 1999, no. 2, pp. 34-37 (Roche Molecular
Biochemicals).
[00288] Apoptosis can also be determined by measuring morphological changes in
a
cell. For example, as with necrosis, loss of plasma membrane integrity can be
determined
by measuring uptake of certain dyes (e.g., a fluorescent dye such as, for
example, acridine
orange or ethidium bromide). A method for measuring apoptotic cell number has
been
described by Duke and Cohen, Current Protocols in Immunology (Coligan et al.
eds.,
1992, pp. 3.17.1-3.17.16). Cells also can be labeled with a DNA dye (e.g.,
acridine
orange, ethidium bromide, or propidium iodide) and the cells observed for
chromatin
condensation and margination along the inner nuclear membrane. Other
morphological
changes that can be measured to determine apoptosis include, e.g., cytoplasmic
condensation, increased membrane blebbing, and cellular shrinkage.

89


CA 02754531 2011-09-02
WO 2010/111018 PCT/US2010/026429
[00289] The presence of apoptotic cells can be measured in both the attached
and
"floating" compartments of the cultures. For example, both compartments can be
collected by removing the supernatant, trypsinizing the attached cells,
combining the
preparations following a centrifugation wash step (e.g., 10 minutes at 2000
rpm), and
detecting apoptosis (e.g., by measuring DNA fragmentation). (See, e.g., Piazza
et al.,
1995, Cancer Research 55:3110-16).
[00290] In vivo, the effect of a 24P4C12 therapeutic composition can be
evaluated in a
suitable animal model. For example, xenogenic cancer models can be used,
wherein
cancer explants or passaged xenograft tissues are introduced into immune
compromised
animals, such as nude or SCID mice (Klein et al., 1997, Nature Medicine 3: 402-
408).
For example, PCT Patent Application W098/16628 and U.S. Patent 6,107,540
describe
various xenograft models of human prostate cancer capable of recapitulating
the
development of primary tumors, micrometastasis, and the formation of
osteoblastic
metastases characteristic of late stage disease. Efficacy can be predicted
using assays that
measure inhibition of tumor formation, tumor regression or metastasis, and the
like.
[00291] In vivo assays that evaluate the promotion of apoptosis are useful in
evaluating
therapeutic compositions. In one embodiment, xenografts from tumor bearing
mice
treated with the therapeutic composition can be examined for the presence of
apoptotic
foci and compared to untreated control xenograft-bearing mice. The extent to
which
apoptotic foci are found in the tumors of the treated mice provides an
indication of the
therapeutic efficacy of the composition.
[00292] The therapeutic compositions used in the practice of the foregoing
methods
can be formulated into pharmaceutical compositions comprising a carrier
suitable for the
desired delivery method. Suitable carriers include any material that when
combined with
the therapeutic composition retains the anti-tumor function of the therapeutic
composition
and is generally non-reactive with the patient's immune system. Examples
include, but
are not limited to, any of a number of standard pharmaceutical carriers such
as sterile
phosphate buffered saline solutions, bacteriostatic water, and the like (see,
generally,
Remington's Pharmaceutical Sciences 16th Edition, A. Osal., Ed., 1980).
[00293] Therapeutic formulations can be solubilized and administered via any
route
capable of delivering the therapeutic composition to the tumor site.
Potentially effective
routes of administration include, but are not limited to, intravenous,
parenteral,



CA 02754531 2011-09-02
WO 2010/111018 PCT/US2010/026429
intraperitoneal, intramuscular, intratumor, intradermal, intraorgan,
orthotopic, and the
like. A preferred formulation for intravenous injection comprises the
therapeutic
composition in a solution of preserved bacteriostatic water, sterile
unpreserved water,
and/or diluted in polyvinylchloride or polyethylene bags containing 0.9%
sterile Sodium
Chloride for Injection, USP. Therapeutic protein preparations can be
lyophilized and
stored as sterile powders, preferably under vacuum, and then reconstituted in
bacteriostatic water (containing for example, benzyl alcohol preservative) or
in sterile
water prior to injection.
[00294] Dosages and administration protocols for the treatment of cancers
using the
foregoing methods will vary with the method and the target cancer, and will
generally
depend on a number of other factors appreciated in the art.

XII.) Treatment of Cancer(s) Expressing 24P4C12
[00295] The identification of 24P4C12 as a protein that is normally expressed
in a
restricted set of tissues, but which is also expressed in cancers such as
those listed in
Table I, opens a number of therapeutic approaches to the treatment of such
cancers.
[00296] Of note, targeted antitumor therapies have been useful even when the
targeted
protein is expressed on normal tissues, even vital normal organ tissues. A
vital organ is
one that is necessary to sustain life, such as the heart or colon. A non-vital
organ is one
that can be removed whereupon the individual is still able to survive.
Examples of non-
vital organs are ovary, breast, and prostate.
[00297] Expression of a target protein in normal tissue, even vital normal
tissue, does
not defeat the utility of a targeting agent for the protein as a therapeutic
for certain tumors
in which the protein is also overexpressed. For example, expression in vital
organs is not
in and of itself detrimental. In addition, organs regarded as dispensible,
such as the
prostate and ovary, can be removed without affecting mortality. Finally, some
vital
organs are not affected by normal organ expression because of an
immunoprivilege.
Immunoprivileged organs are organs that are protected from blood by a blood-
organ
barrier and thus are not accessible to immunotherapy. Examples of
immunoprivileged
organs are the brain and testis.
[00298] Accordingly, therapeutic approaches that inhibit the activity of a
24P4C12
protein are useful for patients suffering from a cancer that expresses
24P4C12. These
91


CA 02754531 2011-09-02
WO 2010/111018 PCT/US2010/026429
therapeutic approaches generally fall into three classes. The first class
modulates
24P4C 12 function as it relates to tumor cell growth leading to inhibition or
retardation of
tumor cell growth or inducing its killing. The second class comprises various
methods
for inhibiting the binding or association of a 24P4C12 protein with its
binding partner or
with other proteins. The third class comprises a variety of methods for
inhibiting the
transcription of a 24P4C 12 gene or translation of 24P4C 12 mRNA.
[00299] Accordingly, Cancer patients can be evaluated for the presence and
level of
24P4C12 expression, preferably using immunohistochemical assessments of tumor
tissue,
quantitative 24P4C 12 imaging, or other techniques that reliably indicate the
presence and
degree of 24P4C12 expression. Immunohistochemical analysis of tumor biopsies
or
surgical specimens is preferred for this purpose. Methods for
immunohistochemical
analysis of tumor tissues are well known in the art.

XIII.) 24P4C12 as a Target for Antibody-based Therapy
[00300] 24P4C 12 is an attractive target for antibody-based therapeutic
strategies. A
number of antibody strategies are known in the art for targeting both
extracellular and
intracellular molecules (see, e.g., complement and ADCC mediated killing as
well as the
use of intrabodies). Because 24P4C12 is expressed by cancer cells of various
lineages
relative to corresponding normal cells, systemic administration of 24P4C12-
immunoreactive compositions are prepared that exhibit excellent sensitivity
without toxic,
non-specific and/or non-target effects caused by binding of the immunoreactive
composition to non-target organs and tissues. Antibodies specifically reactive
with
domains of 24P4C12 are useful to treat 24P4C 12-expres sing cancers
systemically,
preferably as antibody drug conjugates (i.e. ADCs) wherein the conjugate is
with a toxin
or therapeutic agent.
[00301] Those skilled in the art understand that antibodies can be used to
specifically
target and bind immunogenic molecules such as an immunogenic region of a
24P4C12
sequence shown in Figure 1. In addition, skilled artisans understand that it
is routine to
conjugate antibodies to cytotoxic agents (see, e.g., Slevers et al. Blood
93:11 3678-3684
(June 1, 1999)). When cytotoxic and/or therapeutic agents are delivered
directly to cells,
such as by conjugating them to antibodies specific for a molecule expressed by
that cell
92


CA 02754531 2011-09-02
WO 2010/111018 PCT/US2010/026429
(e.g. 24P4C12), the cytotoxic agent will exert its known biological effect
(i.e.
cytotoxicity) on those cells.
[00302] A wide variety of compositions and methods for using antibody-
cytotoxic
agent conjugates to kill cells are known in the art. In the context of
cancers, typical
methods entail administering to an mammal having a tumor a biologically
effective
amount of a conjugate comprising a selected cytotoxic and/or therapeutic agent
linked to
a targeting agent (e.g. a 24P4C12 MAb, preferably Ha5-1(5)2.1) that binds to
an antigen
(e.g. 24P4C12) expressed, accessible to binding or localized on the cell
surfaces. A
typical embodiment is a method of delivering a cytotoxic and/or therapeutic
agent to a
cell expressing 24P4C12, comprising conjugating the cytotoxic agent to an
antibody that
immunospecifically binds to a 24P4C12 epitope, and, exposing the cell to the
antibody
drug conjugate (ADC). Another illustrative embodiment is a method of treating
an
individual suspected of suffering from metastasized cancer, comprising a step
of
administering parenterally to said individual a pharmaceutical composition
comprising a
therapeutically effective amount of an antibody conjugated to a cytotoxic
and/or
therapeutic agent.
[00303] Cancer immunotherapy using 24P4C12 antibodies can be done in
accordance
with various approaches that have been successfully employed in the treatment
of other
types of cancer, including but not limited to colon cancer (Arlen et al.,
1998, Crit. Rev.
Immunol. 18:133-138), multiple myeloma (Ozaki et al., 1997, Blood 90:3179-
3186,
Tsunenari et al., 1997, Blood 90:2437-2444), gastric cancer (Kasprzyk et al.,
1992,
Cancer Res. 52:2771-2776), B-cell lymphoma (Funakoshi et al., 1996, J.
Immunother.
Emphasis Tumor Immunol. 19:93-101), leukemia (Zhong et al., 1996, Leuk. Res.
20:581-
589), colorectal cancer (Moun et al., 1994, Cancer Res. 54:6160-6166; Velders
et al.,
1995, Cancer Res. 55:4398-4403), and breast cancer (Shepard et al., 1991, J.
Clin.
Immunol. 11:117-127). Some therapeutic approaches involve conjugation of naked
antibody to a toxin or radioisotope, such as the conjugation of Y91 or I131 to
anti-CD20
antibodies (e.g., ZevalinTM, IDEC Pharmaceuticals Corp. or BexxarTM, Coulter
Pharmaceuticals) respectively, while others involve co-administration of
antibodies and
other therapeutic agents, such as HerceptinTm (trastuzu MAb) with paclitaxel
(Genentech,
Inc.). In a preferred embodiment, the antibodies will be conjugated a
cytotoxic agent,
supra, preferably an aurastatin derivative designated MMAE (Seattle Genetics).

93


CA 02754531 2011-09-02
WO 2010/111018 PCT/US2010/026429
[00304] Although 24P4C12 antibody therapy is useful for all stages of cancer,
antibody therapy can be particularly appropriate in advanced or metastatic
cancers.
Treatment with the antibody therapy of the invention is indicated for patients
who have
received one or more rounds of chemotherapy. Alternatively, antibody therapy
of the
invention is combined with a chemotherapeutic or radiation regimen for
patients who
have not received chemotherapeutic treatment. Additionally, antibody therapy
can enable
the use of reduced dosages of concomitant chemotherapy, particularly for
patients who do
not tolerate the toxicity of the chemotherapeutic agent very well. Fan et al.
(Cancer Res.
53:4637-4642, 1993), Prewett et al. (International J. of Onco. 9:217-224,
1996), and
Hancock et al. (Cancer Res. 51:4575-4580, 1991) describe the use of various
antibodies
together with chemotherapeutic agents.
[00305] 24P4C12 monoclonal antibodies that treat colon and other cancers
(Table I)
include those that initiate a potent immune response against the tumor or
those that are
directly cytotoxic. In this regard, 24P4C12 monoclonal antibodies (MAbs) can
elicit
tumor cell lysis by either complement-mediated or antibody-dependent cell
cytotoxicity
(ADCC) mechanisms, both of which require an intact Fc portion of the
immunoglobulin
molecule for interaction with effector cell Fc receptor sites on complement
proteins. In
addition, 24P4C12 MAbs that exert a direct biological effect on tumor growth
are useful
to treat cancers that express 24P4C12. Mechanisms by which directly cytotoxic
MAbs
act include: inhibition of cell growth, modulation of cellular
differentiation, modulation
of tumor angiogenesis factor profiles, and the induction of apoptosis. The
mechanism(s)
by which a particular 24P4C12 MAb exerts an anti-tumor effect is evaluated
using any
number of in vitro assays that evaluate cell death such as ADCC, complement-
mediated
cell lysis, and so forth, as is generally known in the art.
[00306] Accordingly, preferred monoclonal antibodies used in the therapeutic
methods
of the invention are those that are either fully human and that bind
specifically to the
target 24P4C 12 antigen with high affinity.

XIV.) 24P4C12 ADC Cocktails
[00307] Therapeutic methods of the invention contemplate the administration of
single
24P4C12 ADCs as well as combinations, or cocktails, of different MAbs (i.e.
24P4C12
MAbs or Mabs that bind another protein). Such MAb cocktails can have certain

94


CA 02754531 2011-09-02
WO 2010/111018 PCT/US2010/026429
advantages inasmuch as they contain MAbs that target different epitopes,
exploit different
effector mechanisms or combine directly cytotoxic MAbs with MAbs that rely on
immune effector functionality. Such MAbs in combination can exhibit
synergistic
therapeutic effects. In addition, 24P4C12 MAbs can be administered
concomitantly with
other therapeutic modalities, including but not limited to various
chemotherapeutic and
biologic agents, androgen-blockers, immune modulators (e.g., IL-2, GM-CSF),
surgery or
radiation. In a preferred embodiment, the 24P4C12 MAbs are administered in
conjugated
form.
[00308] 24P4C12 ADC formulations are administered via any route capable of
delivering the antibodies to a tumor cell. Routes of administration include,
but are not
limited to, intravenous, intraperitoneal, intramuscular, intratumor,
intradermal, and the
like. Treatment generally involves repeated administration of the 24P4C12 ADC
preparation, via an acceptable route of administration such as intravenous
injection (IV),
typically at a dose in the range, including but not limited to, 0.1,
.2,.3,.4,.5,.6,.7,.8,. 9,
1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 15, 20, or 25 mg/kg body weight. In general,
doses in the range
of 10-1000 mg MAb per week are effective and well tolerated.
[00309] Based on clinical experience with the Herceptin (Trastuzumab) in the
treatment of metastatic breast cancer, an initial loading dose of
approximately 4 mg/kg
patient body weight IV, followed by weekly doses of about 2 mg/kg IV of the
MAb
preparation represents an acceptable dosing regimen. Preferably, the initial
loading dose
is administered as a 90-minute or longer infusion. The periodic maintenance
dose is
administered as a 30 minute or longer infusion, provided the initial dose was
well
tolerated. As appreciated by those of skill in the art, various factors can
influence the
ideal dose regimen in a particular case. Such factors include, for example,
the binding
affinity and half life of the MAbs used, the degree of 24P4C12 expression in
the patient,
the extent of circulating shed 24P4C12 antigen, the desired steady-state
antibody
concentration level, frequency of treatment, and the influence of
chemotherapeutic or
other agents used in combination with the treatment method of the invention,
as well as
the health status of a particular patient.
[00310] Optionally, patients should be evaluated for the levels of 24P4C12 in
a given
sample (e.g. the levels of circulating 24P4C12 antigen and/or 24P4C12
expressing cells)
in order to assist in the determination of the most effective dosing regimen,
etc. Such



CA 02754531 2011-09-02
WO 2010/111018 PCT/US2010/026429
evaluations are also used for monitoring purposes throughout therapy, and are
useful to
gauge therapeutic success in combination with the evaluation of other
parameters (for
example, urine cytology and/or ImmunoCyt levels in bladder cancer therapy, or
by
analogy, serum PSA levels in prostate cancer therapy).
[00311] An object of the present invention is to provide 24P4C12 ADCs, which
inhibit
or retard the growth of tumor cells expressing 24P4C12. A further object of
this
invention is to provide methods to inhibit angiogenesis and other biological
functions and
thereby reduce tumor growth in mammals, preferably humans, using such 24P4C 12
ADCs, and in particular using such 24P4C12 ADCs combined with other drugs or
immunologically active treatments.

XV.) Combination Therapy
[00312] In one embodiment, there is synergy when tumors, including human
tumors,
are treated with 24P4C12 ADCs in conjunction with chemotherapeutic agents or
radiation
or combinations thereof. In other words, the inhibition of tumor growth by a
24P4C 12
ADC is enhanced more than expected when combined with chemotherapeutic agents
or
radiation or combinations thereof. Synergy may be shown, for example, by
greater
inhibition of tumor growth with combined treatment than would be expected from
a
treatment of only 24P4C 12 ADC or the additive effect of treatment with a
24P4C 12 ADC
and a chemotherapeutic agent or radiation. Preferably, synergy is demonstrated
by
remission of the cancer where remission is not expected from treatment either
from a
24P4C12 ADC or with treatment using an additive combination of a 24P4C12 ADC
and a
chemotherapeutic agent or radiation.
[00313] The method for inhibiting growth of tumor cells using a 24P4C12 ADC
and a
combination of chemotherapy or radiation or both comprises administering the
24P4C12
ADC before, during, or after commencing chemotherapy or radiation therapy, as
well as
any combination thereof (i.e. before and during, before and after, during and
after, or
before, during, and after commencing the chemotherapy and/or radiation
therapy). For
example, the 24P4C12 ADC is typically administered between 1 and 60 days,
preferably
between 3 and 40 days, more preferably between 5 and 12 days before commencing
radiation therapy and/or chemotherapy. However, depending on the treatment
protocol

96


CA 02754531 2011-09-02
WO 2010/111018 PCT/US2010/026429
and the specific patient needs, the method is performed in a manner that will
provide the
most efficacious treatment and ultimately prolong the life of the patient.
[00314] The administration of chemotherapeutic agents can be accomplished in a
variety of ways including systemically by the parenteral and enteral routes.
In one
embodiment, the 24P4C12 ADCs and the chemotherapeutic agent are administered
as
separate molecules. Particular examples of chemotherapeutic agents or
chemotherapy
include cisplatin, dacarbazine (DTIC), dactinomycin, mechlorethamine (nitrogen
mustard), streptozocin, cyclophosphamide, carmustine (BCNU), lomustine (CCNU),
doxorubicin (adriamycin), daunorubicin, procarbazine, mitomycin, cytarabine,
etoposide,
methotrexate, 5-fluorouracil, vinblastine, vincristine, bleomycin, paclitaxel
(taxol),
docetaxel (taxotere), aldesleukin, asparaginase, busulfan, carboplatin,
cladribine,
dacarbazine, floxuridine, fludarabine, hydroxyurea, ifosfamide, interferon
alpha,
leuprolide, megestrol, melphalan, mercaptopurine, plicamycin, mitotane,
pegaspargase,
pentostatin, pipobroman, plicamycin, streptozocin, tamoxifen, teniposide,
testolactone,
thioguanine, thiotepa, uracil mustard, vinorelbine, chlorambucil, taxol and
combinations
thereof.
[00315] The source of radiation, used in combination with a 24P4C12 ADC, can
be
either external or internal to the patient being treated. When the source is
external to the
patient, the therapy is known as external beam radiation therapy (EBRT). When
the
source of radiation is internal to the patient, the treatment is called
brachytherapy (BT).
[00316] The above described therapeutic regimens may be further combined with
additional cancer treating agents and/or regimes, for example additional
chemotherapy,
cancer vaccines, signal transduction inhibitors, agents useful in treating
abnormal cell
growth or cancer, antibodies (e.g. Anti-CTLA-4 antibodies as described in
WO/2005/092380 (Pfizer)) or other ligands that inhibit tumor growth by binding
to IGF-
1R, and cytokines.
[00317] When the mammal is subjected to additional chemotherapy,
chemotherapeutic
agents described above may be used. Additionally, growth factor inhibitors,
biological
response modifiers, anti-hormonal therapy, selective estrogen receptor
modulators
(SERMs), angiogenesis inhibitors, and anti-androgens may be used. For example,
anti-
hormones, for example anti-estrogens such as Nolvadex (tamoxifen) or, anti-
androgens

97


CA 02754531 2011-09-02
WO 2010/111018 PCT/US2010/026429
such as Casodex (4'-cyano-3-(4-fluorophenylsulphonyl)-2-hydroxy-2-methyl-3-
(trifluoromethyl)propionanilide) may be used.
[00318] The above therapeutic approaches can be combined with any one of a
wide
variety of surgical, chemotherapy or radiation therapy regimens. The
therapeutic
approaches of the invention can enable the use of reduced dosages of
chemotherapy (or
other therapies) and/or less frequent administration, an advantage for all
patients and
particularly for those that do not tolerate the toxicity of the
chemotherapeutic agent well.
XVI.) Kits/Articles of Manufacture
[00319] For use in the laboratory, prognostic, prophylactic, diagnostic and
therapeutic
applications described herein, kits are within the scope of the invention.
Such kits can
comprise a carrier, package, or container that is compartmentalized to receive
one or
more containers such as vials, tubes, and the like, each of the container(s)
comprising one
of the separate elements to be used in the method, along with a label or
insert comprising
instructions for use, such as a use described herein. For example, the
container(s) can
comprise an antibody that is or can be detectably labeled. Kits can comprise a
container
comprising a Drug Unit. The kit can include all or part of the amino acid
sequences in
Figure 2, or Figure 3 or analogs thereof, or a nucleic acid molecule that
encodes such
amino acid sequences.
[00320] The kit of the invention will typically comprise the container
described above
and one or more other containers associated therewith that comprise materials
desirable
from a commercial and user standpoint, including buffers, diluents, filters,
needles,
syringes; carrier, package, container, vial and/or tube labels listing
contents and/or
instructions for use, and package inserts with instructions for use.
[00321] A label can be present on or with the container to indicate that the
composition
is used for a specific therapy or non-therapeutic application, such as a
prognostic,
prophylactic, diagnostic or laboratory application, and can also indicate
directions for
either in vivo or in vitro use, such as those described herein. Directions and
or other
information can also be included on an insert(s) or label(s) which is included
with or on
the kit. The label can be on or associated with the container. A label a can
be on a
container when letters, numbers or other characters forming the label are
molded or
etched into the container itself; a label can be associated with a container
when it is

98


CA 02754531 2011-09-02
WO 2010/111018 PCT/US2010/026429
present within a receptacle or carrier that also holds the container, e.g., as
a package
insert. The label can indicate that the composition is used for diagnosing,
treating,
prophylaxing or prognosing a condition, such as a cancer of a tissue set forth
in Table I.
[00322] The terms "kit" and "article of manufacture" can be used as synonyms.
[00323] In another embodiment of the invention, an article(s) of manufacture
containing compositions, such as antibody(s), or antibody drug conjugates
(ADCs) e.g.,
materials useful for the diagnosis, prognosis, prophylaxis and/or treatment of
cancers of
tissues such as those set forth in Table I is provided. The article of
manufacture typically
comprises at least one container and at least one label. Suitable containers
include, for
example, bottles, vials, syringes, and test tubes. The containers can be
formed from a
variety of materials such as glass, metal or plastic. The container can hold
amino acid
sequence(s), small molecule(s), nucleic acid sequence(s), cell population(s)
and/or
antibody(s). In another embodiment a container comprises an antibody, binding
fragment
thereof or specific binding protein for use in evaluating protein expression
of 24P4C12 in
cells and tissues, or for relevant laboratory, prognostic, diagnostic,
prophylactic and
therapeutic purposes; indications and/or directions for such uses can be
included on or
with such container, as can reagents and other compositions or tools used for
these
purposes.
[00324] The container can alternatively hold a composition that is effective
for
treating, diagnosis, prognosing or prophylaxing a condition and can have a
sterile access
port (for example the container can be an intravenous solution bag or a vial
having a
stopper pierceable by a hypodermic injection needle). The active agents in the
composition can be an antibody capable of specifically binding 24P4C12 or an
antibody
drug conjugate specifically binding to 24P4C12.
[00325] The article of manufacture can further comprise a second container
comprising
a pharmaceutically-acceptable buffer, such as phosphate-buffered saline,
Ringer's solution
and/or dextrose solution. It can further include other materials desirable
from a
commercial and user standpoint, including other buffers, diluents, filters,
stirrers, needles,
syringes, and/or package inserts with indications and/or instructions for use.

99


CA 02754531 2011-09-02
WO 2010/111018 PCT/US2010/026429
EXAMPLES:
[00326] Various aspects of the invention are further described and illustrated
by way of
the several examples that follow, none of which is intended to limit the scope
of the
invention.

Example 1
The 24P4C 12 Antigen

[00327] The novel 24P4C12 gene sequence was discovered using Suppression
Subtractive Hybridization (SSH) methods known in the art. The 24P4C12 SSH
sequence
of 160 bp was identified from a LAPC xenograft SSH experiment using standard
methods. A full length cDNA clone for 24P4C12 was isolated from a LAPC-9 AD
minus benign prostatic hyperplasia experiment. The cDNA is 2587 bp in length
and
encodes a 710 amino acid ORF (See, Figure IA). For further reference see, U.S.
patent
No. 6,943,235 (Agensys, Inc., Santa Monica, CA), U.S. Patent No. 7,220,823
(Agensys,
Inc., Santa Monica, CA), U.S. Patent No. 7,227,008 (Agensys, Inc., Santa
Monica, CA),
and U.S. Patent No. 7,244,827 (Agensys, Inc., Santa Monica, CA). For exemplary
embodiments of the 24P4C12 antigen and variants thereof, see Figure 1.

Example 2
Generation of 24P4C12 Monoclonal Antibodies (MAbs)
[00328] In one embodiment, therapeutic Monoclonal Antibodies ("MAbs") to
24P4C12 and 24P4C12 variants comprise those that react with epitopes specific
for each
protein or specific to sequences in common between the variants that would
bind,
internalize, disrupt or modulate the biological function of 24P4C12 or 24P4C12
variants,
for example, those that would disrupt the interaction with ligands,
substrates, and binding
partners. Immunogens for generation of such MAbs include those designed to
encode or
contain the extracellular domains or the entire 24P4C 12 protein sequence,
regions
predicted to contain functional motifs, and regions of the 24P4C 12 protein
variants
predicted to be antigenic from computer analysis of the amino acid sequence.
Immunogens include peptides and recombinant proteins such as tag5-24P4C12, a
purified
mammalian cell derived His tagged protein. In addition, cells engineered to
express high

100


CA 02754531 2011-09-02
WO 2010/111018 PCT/US2010/026429
levels of 24P4C12, such as RAT1-24P4C12 or 300.19-24P4C12, are used to
immunize
mice.
[00329] MAbs to 24P4C 12 were generated using XenoMouse technology (Amgem
Fremont) wherein the murine heavy and kappa light chain loci have been
inactivated and
a majority of the human heavy and kappa light chain immunoglobulin loci have
been
inserted. The MAb designated Ha5-1(5)2.1 was generated from immunization of
human
y2 producing XenoMice with RAT(E)-24P4C12 cells.
[00330] The 24P4C12 MAb Ha5-1(5)2.1 specifically binds to recombinant 24P4C12
expressing cells (PC3-24P4C12) and multiple cancer cell lines expressing
24P4C12.
[00331] The hybridoma producing an antibody designated Ha5-1(5)2.1 was sent
(via
Federal Express) to the American Type Culture Collection (ATCC), P.O. Box
1549,
Manassas, VA 20108 on 08-August-2007 and assigned Accession numbers PTA-8602.
[00332] DNA coding sequences for 24P4C12 MAb Ha5-1(5)2.1 was determined after
isolating mRNA from the respective hybridoma cells with Trizol reagent (Life
Technologies, Gibco BRL).
[00333] Anti-24P4C12 Ha5-1(5)2.1 heavy and light chain variable nucleic acid
sequences were sequenced from the hybridoma cells using the following
protocol. Ha5-
1(5)2.1 secreting hybridoma cells were lysed with Trizol reagent (Life
Technologies,
Gibco BRL). Total RNA was purified and quantified. First strand cDNAs was
generated
from total RNA with oligo (dT)12-18 priming using the Gibco-BRL Superscript
Preamplification system. First strand cDNA was amplified using human
immunoglobulin
variable heavy chain primers, and human immunoglobulin variable light chain
primers.
PCR products were sequenced and the variable heavy and light chain regions
determined.
[00334] The nucleic acid and amino acid sequences of the variable heavy and
light
chain regions are listed in Figure 2 and Figure 3. Alignment of Ha5-1(5)2.1
MAb to
human Ig germline is set forth in Figure 4A-4B.

Example 3
Expression of Ha5-1(5)2.1 using Recombinant DNA Methods
[00335] To express Ha5-1(5)2.1 MAb recombinantly in transfected cells, Ha5-
1(5)2.1
MAb variable heavy and light chain sequences were cloned upstream of the human
heavy
chain IgG2 and light chain IgK constant regions, respectively. The complete
Ha5-1(5)2.1
101


CA 02754531 2011-09-02
WO 2010/111018 PCT/US2010/026429
MAb human heavy chain and light chain cassettes were cloned downstream of the
CMV
promoter/enhancer in a cloning vector. A polyadenylation site was included
downstream
of the MAb coding sequence. The recombinant Ha5-1(5)2.1 MAb expressing
constructs
were transfected into 293T, Cos and CHO cells. The Ha5-1(5)2.1 MAb secreted
from
recombinant cells was evaluated for binding to cell surface 24P4C12 by flow
cytometry
(Figure 5). PC3-control and PC3-24P4C12 cells were stained with Ha5-1(5)2.1
MAb
from either hybridoma or from CHO cells transfected with Ha5-1(5)2.1 heavy and
light
chain vector constructs.
[00336] Binding was detected by flow cytometry. Results show that the
recombinantly
expressed Ha5-1(5)2.1 expressed in CHO cells binds 24P4C12 similarly to the
Ha5-
1(5)2.1 purfiied from hybridoma (Figure 5).

Example 4
Antibody Drug Conjugation of Ha5-1(5)2.1 MAb

[00337] The Ha5-1(5)2.1 Mab (Figure 2) was conjugated to an auristatin
deravitive
designated MMAE (Formula XI) using a vc (Val-Cit) linker described herein to
create the
antibody drug conjugate (ADC) of the invention designated Ha5-1(5)2.1vcMMAE
using
the following protocols. The conjugation of the vc (Val-Cit) linker to the
MMAE (Seattle
Genetics, Seattle, WA) was completed using the general method set forth in
Table V to
create the cytotoxic vcMMAE (see, US/2006/0074008).
[00338] Next, the antibody drug conjugate (ADC) of the invention designated H5-

1(5)2.1vcMMAE was made using the following protocols.
[00339] Briefly, a 10 mg/mL solution of the Ha5-1(5)2.1 MAb in 20 mM histidine
at
pH 5.2 is added with a 15% volume of 0.5 M Tris at pH 8.8 to adjust the pH of
the
solution to 8.0-8.2. Then, EDTA and sodium chloride are added to 5 mM and 250
mM
final concentration, respectively, in the reaction mixture. The MAb is then
partially
reduced by adding 2.3 molar equivalents of TCEP (relative to moles of MAb) and
then
stirred at 37 C for 3 hours. The partially reduced MAb solution is then cooled
to 22 C
and 5.1 molar equivalents of vcMMAE (relative to moles of antibody) are added
as a 7
mg/mL solution in DMSO. The mixture is stirred for 30 minutes at 22 C, then
for 15
additional minutes following the addition of 2 molar equivalents of N-
acetylcysteine
relative to vcMMAE. Excess quenched vcMMAE and other reaction components are

102


CA 02754531 2011-09-02
WO 2010/111018 PCT/US2010/026429
removed by ultrafiltration/diafiltration of the antibody drug conjugate (ADC)
with 10
diavolumes of 20 mM histidine, pH 5.2.
[00340] The resulting antibody drug conjugate (ADC) is designated Ha5-
1(5)2.1vcMMAE and has the following formula:

H3C
O H CH3 H OH
N OJ`N N N'~ ^ II!J N
O
N j,, 0 OCH30 OCH30
N II .40
mAb 3 0 0

NH
O=(
[00341] NH2
Wherein MAb is Ha5-1(5)2.1 (Figure 2 and Figure 3) and p is from 1 to 8. The p
value of
the antibody drug conjugate set forth in this Example was about 3.6.

Example 5
Characterization of HA5-1(5)2.1vcMMAE

[00342] Antibody Drug Conjugates that bind 24P4C12 were generated using the
procedures set forth in the example entitled "Antibody Drug Conjugation of Ha5-
1(5)2.1
MAb" and were screened, identified, and characterized using a combination of
assays
known in the art.
A. Affinity Determination by FACS
[00343] Ha5-1(5)2.1vcMMAE was tested for its binding affinity to 24P4C12
endogenously expressed on LNCaP cells. Briefly, eleven (11) dilutions of Ha5-
1(5)2.1vcMMAE are incubated with LNCaP cells (50,000 cells per well) overnight
at 4 C
at a final concentration of 160 nM to 0.011 nM. At the end of the incubation,
cells are
washed and incubated with anti-hIgG-PE detection antibody for 45 min at 4 C.
After
washing the unbound detection antibodies, the cells are analyzed by FACS. Mean
Florescence Intensity (MFI) values were obtained as listed in (Table IV(A)).
MFI values
were entered into Graphpad Prisim software and analyzed using the one site
binding
(hyperbola) equation of Y=Bmax*X/(Kd+X) to generate Ha5-1(5)2.1vcMMAE
saturation
curves shown in (Table IV(B)). Bmax is the MFI value at maximal binding of Ha5-


103


CA 02754531 2011-09-02
WO 2010/111018 PCT/US2010/026429
1(5)2.1vcMMAE to 24P4C12; Kd is Ha5-1(5)2.1vcMMAE binding affinity which is
the
concentration of Ha5-1(5)2.1vcMMAE required to reach half-maximal binding.
[00344] The calculated affinity (Kd) of Ha5-1(5)2.1vcMMAE is 1.05 nM on
24P4C12
endogenously expressed on the surface of LNCaP cells.

Example 6
Cell Cyootoxicity Mediated by Ha5-1(5)2.1vcMMAE

[00345] The ability of Ha5-1(5)2.1-vcMMAE to mediate 24P4C12-dependent
cytoxicity was evaluated in PC3 cells engineered to express 24P4C12. PC3-Neo
or PC3-
24P4C12 cells (1000 cells/well) were seeded into a 96 well plate on day 1. The
following
day an equal volume of medium containing the indicated concentration of Ha5-
1(5)2.1-
vcMMAE or a Control MAb conjugated with vc-MMAE (i.e. Control-vcMMAE) was
added to each well. The cells were allowed to incubate for 4 days at 37
degrees C. At the
end of the incubation period, Alamar Blue was added to each well and
incubation
continued for an additional 4 hours. The resulting fluorescence was detected
using a
Biotek plate reader with an excitation wavelength of 620 nm and an emission
wavelength
of 540 nm.
[00346] The results in Figure 6 show that Ha5-1(5)2.1-vcMMAE mediated
cytotoxicity in PC3-24P4C12 cells while a control human IgG conjugated with
vcMMAE
had no effect. The specificity of Ha5-1(5)2.1-vcMMAE was further demonstrated
by the
lack of toxicity for PC3-Neo cells that do not express 24P4C12. Thus, these
results
indicate that Ha5-1(5)2.1-vcMMAE can selectively deliver a cytotoxic drug to
24P4C12
expressing cells leading to their killing.

Example 7
Ha5-1(5)2.1vcMMAE Inhibit Growth of Tumors In Vivo

[00347] The significant expression of 24P4C12 on the cell surface of tumor
tissues,
together with its restrictive expression in normal tissues makes 24P4C12 a
good target for
antibody therapy and similarly, therapy via ADC. Thus, the therapeutic
efficacy of Ha5-
1(5)2.1vcMMAE in human ovarian, prostate, colon, and pancreatic cancer
xenograft
mouse models is evaluated.

104


CA 02754531 2011-09-02
WO 2010/111018 PCT/US2010/026429
[00348] Antibody drug conjugate efficacy on tumor growth and metastasis
formation
is studied in mouse cancer xenograft models (e.g. subcutaneous and
orthotopically).
[00349] Subcutaneous (s.c.) tumors are generated by injection of 5 x 104- 106
cancer
cells mixed at a 1:1 dilution with Matrigel (Collaborative Research) in the
right flank of
male SCID mice. To test ADC efficacy on tumor formation, i.e. ADC injections
are
started on the same day as tumor-cell injections. As a control, mice are
injected with
either purified human IgG or PBS; or a purified MAb that recognizes an
irrelevant
antigen not expressed in human cells. In preliminary studies, no difference is
found
between control IgG or PBS on tumor growth. Tumor sizes are determined by
caliper
measurements, and the tumor volume is calculated as length x width x height.
Mice with
subcutaneous tumors greater than 1.5 cm in diameter are sacrificed.
[00350] Ovarian tumors often metastasize and grow within the peritoneal
cavity.
Accordingly, intraperitoneal growth of ovarian tumors in mice are performed by
injection of 2 million cells directly into the peritoneum of female mice. Mice
are
monitored for general health, physical activity, and appearance until they
become
moribund. At the time of sacrifice, the peritoneal cavity can be examined to
determine
tumor burden and lungs harvested to evaluate metastasis to distant sites.
Alternatively,
death can be used as an endpoint. The mice are then segregated into groups for
the
appropriate treatments, with 24P4C12 or control MAbs being injected i.p.
[00351] An advantage of xenograft cancer models is the ability to study
neovascularization and angiogenesis. Tumor growth is partly dependent on new
blood
vessel development. Although the capillary system and developing blood network
is of
host origin, the initiation and architecture of the neovasculature is
regulated by the
xenograft tumor (Davidoff et al., Clin Cancer Res. (2001) 7:2870; Solesvik et
al., Eur J
Cancer Clin Oncol. (1984) 20:1295). The effect of antibody and small molecule
on
neovascularization is studied in accordance with procedures known in the art,
such as by
IHC analysis of tumor tissues and their surrounding microenvironment.
[00352] Ha5-1(5)2.1ADC inhibits formation colon, pancreatic, ovarian, and
prostate
cancer xenografts. These results indicate the utility of Ha5-1(5)2.1ADC in the
treatment
of local and advanced stages of cancer and preferably those cancers set forth
in Table I.
24P4C12 ADCs:

105


CA 02754531 2011-09-02
WO 2010/111018 PCT/US2010/026429
[00353] Monoclonal antibodies were raised against 24P4C12 as described in the
Example entitled "Generation of 24P4C12 Monoclonal Antibodies (MAbs)." Further
the
MAbs are conjugated to a toxin as described in the Example entitled "Antibody
Drug
Conjugation of Ha5-1(5)2.1 MAb" to form AGS-5M2.1vcMMAE. The Ha5-
1(5)2.1vcMMAE is characterized by FACS, and other methods known in the art to
determine its capacity to bind 24P4C12.

Cell Lines and Xenografts:
[00354] The PC3-24P4C12, LAPC9, HT-29, AG-C4, OVCAR5-24P4C12, and AG-
Panc3 cells are maintained in DMEM and RPMI respectively, supplemented with L-
glutamine and 10% FBS. LAPC9, AG-C4, and AG-PAnc3 xenografts are maintained by
serial propogation in SCID mice.

Ha5-1(5)2.1vcMMAE inhibits the Growth of Subcutaneous established human
androgen-independent prostate cancer xenograft in SCID mice
[00355] In this experiment, androgen-independent human prostate cancer PC3-
24P4C12 tumor cells (3.0 x 106 cells/mouse) were injected subcutaneously into
male
SCID mice. Mice were randomized into Ha5-1(5)2.1-vcMMAE and PBS control groups
(n=5 in each group) when tumors reached 100 mm3. Mice were treated with a
single dose
of Ha5-1(5)2.1-vcMMAE (10 mg/kg) or PBS administered intravenously (i.v.) on
Day 0.
Tumor growth was monitored using caliper measurements every 3 to 4 days as
indicated.
Tumor volume was calculated as Width2 x Length/2, where width is the smallest
dimension and length is the largest.
[00356] The results show that treatment with Ha5-1(5)2.1-vcMMAE significantly
inhibited the growth of PC-3-Hu24P4C12 prostate tumors in SCID mice (p< 0.01)
and
resulted in complete tumor regression in most animals. (Figure 7)

Ha5-1(5)2.1vcMMAE inhibits the Growth of Orthotopically established human
androgen-independent prostate cancer xenograft in SCID mice
[00357] In another experiment, LAPC-9AI androgen-independent human prostate
cancer cells (2.0 x 106 cells/mouse) were implanted into the prostates of male
SCID mice.
Fifteen (15) days after implantation when tumors were well established and
palpable, the
106


CA 02754531 2011-09-02
WO 2010/111018 PCT/US2010/026429
mice were randomized into two groups (n=8 in each group). Mice were treated
with either
Ha5-1(5)2.1-vcMMAE or isotype control MAb conjugated with vcMMAE administered
i.v. at 3 mg/kg every 4 days for a total of 4 doses. At the end of study
tumors in the
mouse prostate were excised and weighed using an electronic balance.
[00358] The results show that treatment with Ha5-1(5)2.1-vcMMAE significantly
inhibited the growth of LAPC9-AI human prostate tumors implanted
orthotopically in
SCID mice (p< 0.01). (Figure 8).

Ha5-1(5)2.1vcMMAE inhibits the Growth of Subcutaneous established human
androgen-independent human colon cancer xenograft in SCID mice
[00359] In another experiment, HT-29 human colon cancer cells (1.0 x 106
cells/mouse) were injected subcutaneously into SCID mice. Mice were randomized
into
two groups (n=6 in each group) when tumors reached 100 mm3. Ha5-1(5)2.1-vcMMAE
(3 mg/kg) or PBS was administered intravenously every 4 days for a total of 4
doses
beginning on Day 0. Tumor growth was monitored using caliper measurements
every 3 to
4 days as indicated. Tumor volume was calculated as Width2 x Length/2, where
width is
the smallest dimension and length is the largest.
[00360] The results show that treatment with Ha5-1(5)2.1-vcMMAE significantly
inhibited the growth of HT-29 human colon tumor xenografts implanted
subcutaneously
in SCID mice (p< 0.01). (Figure 9).

Ha5-1(5)2.1vcMMAE inhibits the Growth of Subcutaneous established human
androgen-independent patient-derived colon cancer xenograft in SCID mice
[00361] In another experiment, AG-C4, patient-derived colon cancer xenograft
tumor
pieces, were implanted subcutaneously into SCID mice. Mice were randomized
into two
groups (n=6 in each group) when tumors reached 100 mm3. Ha5-1(5)2.1-vcMMAE (3
mg/kg ) or PBS was administered intravenously every 3-4 days for a total of 4
doses
starting on Day 0. Tumor growth was monitored using caliper measurements every
3 to 4
days as indicated. Tumor volume was calculated as Width2 x Length/2, where
width is the
smallest dimension and length is the largest.

107


CA 02754531 2011-09-02
WO 2010/111018 PCT/US2010/026429
[00362] The results show that treatment with Ha5-1(5)2.1-vcMMAE significantly
inhibited the growth of AG-C4 human colon tumor xenografts implanted
subcutaneously
in SCID mice (p< 0.05). (Figure 10).

Ha5-1(5)2.1vcMMAE inhibits the Growth of Subcutaneous established human
ovarian cancer xenograft in nude mice
[00363] In another experiment, OVCAR-5 human ovarian cancer tumor cells (2.0 x
106 cells/mouse) were injected subcutaneously into the nude mice. Mice were
randomized
into two groups (n=6 in each group) when tumors reached 100 mm3. Ha5-1(5)2.1-
vcMMAE (5 mg/kg) or PBS was administered intravenously once every 3-4 days for
a
total of 4 doses starting on Day 0. Tumor growth was monitored using caliper
measurements every 3 to 4 days as indicated. Tumor volume was calculated as
Width2 x
Length/2, where width is the smallest dimension and length is the largest.
[00364] The results show that treatment with Ha5-1(5)2.1-vcMMAE significantly
inhibited the growth of OVCAR-5 ovarian cancer xenografts implanted
subcutaneously in
nude mice (p< 0.01). (Figure 11).

Ha5-1(5)2.1vcMMAE inhibits the Growth of Subcutaneous established patient-
derived pancreatic cancer xenograft in SCID mice
[00365] In this experiment, AG-Panc3 patient-derived pancreatic tumor pieces
were
implanted subcutaneously into SCID mice. Mice were randomized into two groups
(n=6
in each group) when tumors reached 85 mm3. Ha5-1(5)2.1-vcMMAE (5 mg/kg) or PBS
was administered intravenously once every 3-4 days for a total of 4 doses
beginning on
Day 0. Tumor growth was monitored using caliper measurements every 3 to 4 days
as
indicated. Tumor volume was calculated as Width2 x Length/2, where width is
the
smallest dimension and length is the largest.
[00366] The results show that treatment with Ha5-1(5)2.1-vcMMAE significantly
inhibited the growth of AG-Panc3 tumor xenografts implanted subcutaneously in
SCID
mice (p< 0.01). (Figure 12).
[00367] The results of these experiments show that 24P4C12 ADC designated Ha5-
1(5)2.1vcMMAE can be used for therapeutic purposes to treat and manage cancers
set
forth in Table I.

108


CA 02754531 2011-09-02
WO 2010/111018 PCT/US2010/026429
Efficacy of Ha5-1(5)2.1vcMMAE compared to other 24P4C12 Antibody Drug
Conjugates (ADCs) in Prostate Cancer LAPC9-AD Xeno rg afts
[00368] In another experiment, LAPC-9AD androgen-dependent human prostate
cancer cells (1.5 x 106 cells/mouse) were injected subcutaneously into male
SCID mice.
Mice were randomized into Ha5-1(5)2.1-vcMMAE, Ha5-1(5)2.1-mcMMAF and other
Antibody Drug Conjugate (ADC) groups including a PBS control group (n=6 in
each
group), as shown in graph (Figure 13). When tumors reached 100 mm3, Ha5-
1(5)2.1-
vcMMAE, Ha5-1(5)2.1-mcMMAF and all other ADCs were administered intravenously
at 10 mg/kg once on day 0. Tumor growth was monitored using caliper
measurements
every 3 to 4 days as indicated. Tumor volume was calculated as Width2 x
Length/2, where
width is the smallest dimension and length is the largest.
[00369] The results show that treatment with Ha5-1(5)2.1-vcMMAE significantly
inhibited the growth of LAPC9-AD prostate cancer xenografts as compared to Ha5-

1(5)2.1-mcMMAF (p=0.0048). (Figure 13). Other antibodies conjugated to -vcMMAE
and -mcMMAF did not have any tumor inhibitory activity which shows that Ha5-
1(5)2.1
posesses a significant prominent effect of inhibiting tumor growth and can be
used for
therapeutic purposes to treat and manage cancers set forth in Table I.
Example 8
Human Clinical Trials for the Treatment and Diagnosis of Human Carcinomas
through
use of 24P4C12 ADCs
[00370] 24P4C12 ADCs are used in accordance with the present invention which
specifically bind to 24P4C12, and are used in the treatment of certain tumors,
preferably
those listed in Table I. In connection with each of these indications, two
clinical
approaches are successfully pursued.
[00371] I.) Adjunctive therapy: In adjunctive therapy, patients are treated
with
24P4C12 ADCs in combination with a chemotherapeutic or anti-neoplastic agent
and/or
radiation therapy or a combination thereof. Primary cancer targets, such as
those listed in
Table I, are treated under standard protocols by the addition of 24P4C12 ADCs
to
standard first and second line therapy. Protocol designs address effectiveness
as assessed
by the following examples, including but not limited to, reduction in tumor
mass of
primary or metastatic lesions, increased progression free survival, overall
survival,

109


CA 02754531 2011-09-02
WO 2010/111018 PCT/US2010/026429
improvement of patients health, disease stabilization, as well as the ability
to reduce usual
doses of standard chemotherapy and other biologic agents. These dosage
reductions
allow additional and/or prolonged therapy by reducing dose-related toxicity of
the
chemotherapeutic or biologic agent. 24P4C12 ADCs are utilized in several
adjunctive
clinical trials in combination with the chemotherapeutic or anti-neoplastic
agents.
[00372] IL) Monotherapy: In connection with the use of the 24P4C12 ADCs in
monotherapy of tumors, the 24P4C12 ADCs are administered to patients without a
chemotherapeutic or anti-neoplastic agent. In one embodiment, monotherapy is
conducted clinically in end-stage cancer patients with extensive metastatic
disease.
Protocol designs address effectiveness as assessed by the following examples,
including
but not limited to, reduction in tumor mass of primary or metastatic lesions,
increased
progression free survival, overall survival, improvement of patients health,
disease
stabilization, as well as the ability to reduce usual doses of standard
chemotherapy and
other biologic agents.

Dosage
[00373] Dosage regimens may be adjusted to provide the optimum desired
response.
For example, a single bolus may be administered, several divided doses may be
administered over time or the dose may be proportionally reduced or increased
as
indicated by the exigencies of the therapeutic situation. It is especially
advantageous to
formulate parenteral compositions in dosage unit form for ease of
administration and
uniformity of dosage. Dosage unit form as used herein refers to physically
discrete units
suited as unitary dosages for the mammalian subjects to be treated; each unit
containing a
predetermined quantity of active compound calculated to produce the desired
therapeutic
effect in association with the required pharmaceutical carrier. The
specification for the
dosage unit forms of the invention are dictated by and directly dependent on
(a) the
unique characteristics of the antibody and the particular therapeutic or
prophylactic effect
to be achieved, and (b) the limitations inherent in the art of compounding
such an active
compound for the treatment of sensitivity in individuals.
[00374] An exemplary, non limiting range for a therapeutically effective
amount of an
24P4C12 ADC administered in combination according to the invention is about
0.5 to
about 10 mg/kg, about 1 to about 5 mg/kg, at least 1 mg/kg, at least 2 mg/kg,
at least 3
mg/kg, or at least 4 mg/kg. Other exemplary non-limiting ranges are for
example about
110


CA 02754531 2011-09-02
WO 2010/111018 PCT/US2010/026429
0.5 to about 5 mg/kg, or for example about 0.8 to about 5 mg/kg, or for
example about 1
to about 7.5mg/kg. The high dose embodiment of the invention relates to a
dosage of
more than 10 mg/kg. It is to be noted that dosage values may vary with the
type and
severity of the condition to be alleviated, and may include single or multiple
doses. It is to
be further understood that for any particular subject, specific dosage
regimens should be
adjusted over time according to the individual need and the professional
judgment of the
person administering or supervising the administration of the compositions,
and that
dosage ranges set forth herein are exemplary only and are not intended to
limit the scope
or practice of the claimed composition.
Clinical Development Plan (CDP)
[00375] The CDP follows and develops treatments of 24P4C12 ADcs in connection
with adjunctive therapy or monotherapy. Trials initially demonstrate safety
and thereafter
confirm efficacy in repeat doses. Trials are open label comparing standard
chemotherapy
with standard therapy plus 24P4C12 ADCs. As will be appreciated, one non-
limiting
criteria that can be utilized in connection with enrollment of patients is
24P4C12
expression levels in their tumors as determined by biopsy.
[00376] As with any protein or antibody infusion-based therapeutic, safety
concerns
are related primarily to (i) cytokine release syndrome, i.e., hypotension,
fever, shaking,
chills; (ii) the development of an immunogenic response to the material (i.e.,
development
of human antibodies by the patient to the antibody therapeutic, or HAHA
response); and,
(iii) toxicity to normal cells that express 24P4C12. Standard tests and follow-
up are
utilized to monitor each of these safety concerns. 24P4C12 MAbs are found to
be safe
upon human administration.

Example 9
Detection of 24P4C12 protein in gastric cancer patient specimens by IHC
[00377] Expression of 24P4C12 protein by immunohistochemistry was tested in
two
(2) different tumor specimens from gastric cancer patients. Briefly, formalin
fixed,
paraffin wax-embedded tissues were cut into 4 micron sections and mounted on
glass
slides. The sections were de-waxed, rehydrated and treated with trypsin
solution (0.05%
trypsin (ICN, Aurora, Ohio) in 0.05% calcium chloride, with pH adjusted to
7.8) at 37 C
for 10 minutes. Sections were then treated with 3% hydrogen peroxide solution
to

111


CA 02754531 2011-09-02
WO 2010/111018 PCT/US2010/026429
inactivate endogenous peroxidase activity. Serum-free protein block (Dako,
Carpenteria,
CA) was used to inhibit non-specific binding prior to incubation with
monoclonal mouse
anti-24P4C12 antibody or an isotype control. Subsequently, the sections were
treated
with the Super SensitiveTM Polymer-horseradish peroxidase (HRP) Detection
System
which consists of an incubation in Super EnhancerTM reagent followed by an
incubation
with polymer-HRP secondary antibody conjugate (BioGenex, San Ramon, CA). The
sections were then developed using the DAB kit (BioGenex, San Ramon, CA),
nuclei
were stained using hematoxylin, and analyzed by bright field microscopy.
Specific
staining was detected in patient specimens using the 24P4C12 immunoreactive
antibody,
as indicated by the brown staining. (See, Figures 14(A) and 14(C). In
contrast, the
control antibody did not stain either patient specimen. (See, Figures 14(B)
and 14(D).
The results show expression of 24P4C12 in the tumor cells of patient gastric
cancer
tissues. These results indicate that 24P4C12 is expressed in human cancers and
that
antibodies directed to this antigen (e.g. Ha5-1(5)2.1) are useful for
diagnostic and
therapeutic purposes. (Figure 14(A) - 14(D)).

[00378] Throughout this application, various website data content,
publications, patent
applications and patents are referenced. (Websites are referenced by their
Uniform
Resource Locator, or URL, addresses on the World Wide Web.) The disclosures of
each
of these references are hereby incorporated by reference herein in their
entireties.
[00379] The present invention is not to be limited in scope by the embodiments
disclosed herein, which are intended as single illustrations of individual
aspects of the
invention, and any that are functionally equivalent are within the scope of
the invention.
Various modifications to the models and methods of the invention, in addition
to those
described herein, will become apparent to those skilled in the art from the
foregoing
description and teachings, and are similarly intended to fall within the scope
of the
invention. Such modifications or other embodiments can be practiced without
departing
from the true scope and spirit of the invention.

112


CA 02754531 2011-09-02
WO 2010/111018 PCT/US2010/026429

Tables
Table I: Tissues that express 24P4C12 when malignant.
Colon
Pancreas
Ovarian
Breast
Lung
Prostate
Gastric
TABLE II: Amino Acid Abbreviations

SINGLE LETTER THREE LETTER FULL NAME
F Phe phenylalanine
L Leu leucine
S Ser serine
Y Tyr tyrosine
C Cys c steine
W Trp tr ptophan
P Pro proline
H His histidine
Q Gln lutamine
R Arg arginine
I Ile isoleucine
M Met methionine
T Thr threonine
N Asn as ara ine
K Lys lysine
V Val valine
A Ala alanine
D Asp aspartic acid
E Glu glutamic acid
G Gly glycine

113


CA 02754531 2011-09-02
WO 2010/111018 PCT/US2010/026429
TABLE III: Amino Acid Substitution Matrix

Adapted from the GCG Software 9.0 BLOSUM62 amino acid substitution matrix
(block substitution matrix). The higher
the value, the more likely a substitution is found in related, natural
proteins.

A C D E F G H I K L M N P Q R S T V W Y.
4 0 -2 -1 -2 0 -2 -1 -1 -1 -1 -2 -1 -1 -1 1 0 0 -3 -2 A
9 -3 -4 -2 -3 -3 -1 -3 -1 -1 -3 -3 -3 -3 -1 -1 -1 -2 -2 C
6 2 -3 -1 -1 -3 -1 -4 -3 1 -1 0 -2 0 -1 -3 -4 -3 D
-3 -2 0 -3 1 -3 -2 0 -1 2 0 0 -1 -2 -3 -2 E
6 -3 -1 0 -3 0 0 -3 -4 -3 -3 -2 -2 -1 1 3 F
6 -2 -4 -2 -4 -3 0 -2 -2 -2 0 -2 -3 -2 -3 G
8 -3 -1 -3 -2 1 -2 0 0 -1 -2 -3 -2 2 H
4 -3 2 1 -3 -3 -3 -3 -2 -1 3 -3 -1 I
5 -2 -1 0 -1 1 2 0 -1 -2 -3 -2 K
4 2 -3 -3 -2 -2 -2 -1 1 -2 -1 L
5 -2 -2 0 -1 -1 -1 1 -1 -1 M
6 -2 0 0 1 0 -3 -4 -2 N
7 -1 -2 -1 -1 -2 -4 -3 P
5 1 0 -1 -2 -2 -1 Q
5 -1 -1 -3 -3 -2 R
4 1 -2 -3 -2 S
5 0 -2 -2 T
4 -3 -1 V
11 2 W
7 Y
114


CA 02754531 2011-09-02
WO 2010/111018 PCT/US2010/026429
Table IV A : FACS MFI of AGS-5M2.1vcMMAE on LnCAP cells

Ha5- MFI on
1(5)2.1 vcMMAE LNCaP
Conc. (nM) cells
160.000 116
106.667 114
71.111 108
23.704 97
7.901 86
2.634 70
0.878 54
0.293 28
0.098 15
0.033 9
0.011 7

Table IV B : Affinity values calculated by GraphPad Prisim software
AGS-5M2.1vcMMAE saturation curve

LnCAP cells
12
^ =
^

75 ^ AGS-5M2ADC
5


0 25 50 75 100 125 150 175
AGS-5M2ADC Conc. (nM)

115


CA 02754531 2011-09-02
WO 2010/111018 PCT/US2010/026429
Table V. General Method for Synthesis of vcMMAE

Where: AA1 = Amino Acid 1
AA2 = Amino Acid 2
AA5 = Amino Acid 5
DIL = Dolaisoleuine
DAP = Dolaproine
Linker = Val-Cit (vc)

OtBu
r-ry
I.~:.~ OCH
~;p0\ 3

... ... QN OH
Bod
Y OCH3 0
Dap

') \ - A&
1
--

116

Representative Drawing

Sorry, the representative drawing for patent document number 2754531 was not found.

Administrative Status

For a clearer understanding of the status of the application/patent presented on this page, the site Disclaimer , as well as the definitions for Patent , Administrative Status , Maintenance Fee  and Payment History  should be consulted.

Administrative Status

Title Date
Forecasted Issue Date Unavailable
(86) PCT Filing Date 2010-03-05
(87) PCT Publication Date 2010-09-30
(85) National Entry 2011-09-02
Dead Application 2016-03-07

Abandonment History

Abandonment Date Reason Reinstatement Date
2015-03-05 FAILURE TO REQUEST EXAMINATION
2015-03-05 FAILURE TO PAY APPLICATION MAINTENANCE FEE

Payment History

Fee Type Anniversary Year Due Date Amount Paid Paid Date
Application Fee $400.00 2011-09-02
Maintenance Fee - Application - New Act 2 2012-03-05 $100.00 2012-02-21
Maintenance Fee - Application - New Act 3 2013-03-05 $100.00 2013-02-22
Maintenance Fee - Application - New Act 4 2014-03-05 $100.00 2014-02-20
Owners on Record

Note: Records showing the ownership history in alphabetical order.

Current Owners on Record
SEATTLE GENETICS, INC.
AGENSYS, INC.
Past Owners on Record
None
Past Owners that do not appear in the "Owners on Record" listing will appear in other documentation within the application.
Documents

To view selected files, please enter reCAPTCHA code :



To view images, click a link in the Document Description column. To download the documents, select one or more checkboxes in the first column and then click the "Download Selected in PDF format (Zip Archive)" or the "Download Selected as Single PDF" button.

List of published and non-published patent-specific documents on the CPD .

If you have any difficulty accessing content, you can call the Client Service Centre at 1-866-997-1936 or send them an e-mail at CIPO Client Service Centre.


Document
Description 
Date
(yyyy-mm-dd) 
Number of pages   Size of Image (KB) 
Abstract 2011-09-02 1 67
Claims 2011-09-02 2 62
Drawings 2011-09-02 37 1,810
Description 2011-09-02 116 5,387
Cover Page 2011-11-07 2 34
PCT 2011-09-02 11 456
Assignment 2011-09-02 2 74
Prosecution-Amendment 2011-09-02 1 15
Prosecution-Amendment 2011-12-01 66 2,441
Correspondence 2012-03-16 3 97

Biological Sequence Listings

Choose a BSL submission then click the "Download BSL" button to download the file.

If you have any difficulty accessing content, you can call the Client Service Centre at 1-866-997-1936 or send them an e-mail at CIPO Client Service Centre.

Please note that files with extensions .pep and .seq that were created by CIPO as working files might be incomplete and are not to be considered official communication.

BSL Files

To view selected files, please enter reCAPTCHA code :